US20040180416A1 - Novel human kinases and polynucleotides encoding the same - Google Patents

Novel human kinases and polynucleotides encoding the same Download PDF

Info

Publication number
US20040180416A1
US20040180416A1 US10/803,277 US80327704A US2004180416A1 US 20040180416 A1 US20040180416 A1 US 20040180416A1 US 80327704 A US80327704 A US 80327704A US 2004180416 A1 US2004180416 A1 US 2004180416A1
Authority
US
United States
Prior art keywords
ser
pro
leu
nhp
arg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/803,277
Other versions
US6919192B2 (en
Inventor
C. Turner
Brian Mathur
Carl Friddle
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/803,277 priority Critical patent/US6919192B2/en
Publication of US20040180416A1 publication Critical patent/US20040180416A1/en
Priority to US11/165,424 priority patent/US20080050809A1/en
Application granted granted Critical
Publication of US6919192B2 publication Critical patent/US6919192B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1205Phosphotransferases with an alcohol group as acceptor (2.7.1), e.g. protein kinases

Definitions

  • the present invention relates to the discovery, identification, and characterization of novel human polynucleotides encoding proteins sharing sequence similarity with animal kinases.
  • the invention encompasses the described polynucleotides, host cell expression systems, the encoded proteins, fusion proteins, polypeptides and peptides, antibodies to the encoded proteins and peptides, and genetically engineered animals that either lack or overexpress the disclosed polynucleotides, antagonists and agonists of the proteins, and other compounds that modulate the expression or activity of the proteins encoded by the disclosed polynucleotides, which can be used for diagnosis, drug screening, clinical trial monitoring, the treatment of diseases and disorders, and cosmetic or nutriceutical applications.
  • kinases mediate the phosphorylation of a wide variety of proteins and compounds in the cell. Along with phosphatases, kinases are involved in a range of regulatory pathways. Given the physiological importance of kinases, they have been subject to intense scrutiny and are proven drug targets.
  • the present invention relates to the discovery, identification, and characterization of nucleotides that encode novel human proteins and the corresponding amino acid sequences of these proteins.
  • novel human proteins (NHPs), described for the first time herein, share structural similarity with animal kinases, including, but not limited to, serine-threonine kinases, and carbon catabolite depressing kinases. Accordingly, the described NHPs encode novel kinases having homologues and orthologs across a range of phyla and species.
  • novel human polynucleotides described herein encode open reading frames (ORFs) encoding proteins of 778, 762, and 703 amino acids in length (see respectively SEQ ID NOS:2, 4, and 6).
  • the invention also encompasses agonists and antagonists of the described NHPs, including small molecules, large molecules, mutant NHPs, or portions thereof, that compete with native NHPs, peptides, and antibodies, as well as nucleotide sequences that can be used to inhibit the expression of the described NHPs (e.g., antisense and ribozyme molecules, and open reading frame or regulatory sequence replacement constructs) or to enhance the expression of the described NHPs (e.g., expression constructs that place the described polynucleotide under the control of a strong promoter system), and transgenic animals that express a NHP sequence, or “knock-outs” (which can be conditional) that do not express a functional NHP.
  • nucleotide sequences that can be used to inhibit the expression of the described NHPs (e.g., antisense and ribozyme molecules, and open reading frame or regulatory sequence replacement constructs) or to enhance the expression of the described NHPs (e.g., expression constructs that
  • Knock-out mice can be produced in several ways, one of which involves the use of mouse embryonic stem cell (“ES cell”) lines that contain gene trap mutations in a murine homolog of at least one of the described NHPs.
  • ES cell mouse embryonic stem cell
  • the unique NHP sequences described in SEQ ID NOS:1-6 are “knocked-out” they provide a method of identifying phenotypic expression of the particular gene, as well as a method of assigning function to previously unknown genes.
  • animals in which the unique NHP sequences described in SEQ ID NOS:1-6 are “knocked-out” provide a unique source in which to elicit antibodies to homologous and orthologous proteins, which would have been previously viewed by the immune system as “self” and therefore would have failed to elicit significant antibody responses.
  • gene trapped knockout ES cells have been generated in murine homologs of the described NHPs.
  • the unique NHP sequences described in SEQ ID NOS:1-6 are useful for the identification of protein coding sequences, and mapping a unique gene to a particular chromosome. These sequences identify biologically verified exon splice junctions, as opposed to splice junctions that may have been bioinformatically predicted from genomic sequence alone.
  • the sequences of the present invention are also useful as additional DNA markers for restriction fragment length polymorphism (RFLP) analysis, and in forensic biology.
  • RFLP restriction fragment length polymorphism
  • the present invention also relates to processes for identifying compounds that modulate, i.e., act as agonists or antagonists of, NHP expression and/or NHP activity that utilize purified preparations of the described NHPs and/or NHP products, or cells expressing the same.
  • Such compounds can be used as therapeutic agents for the treatment of any of a wide variety of symptoms associated with biological disorders or imbalances.
  • the NHPs described for the first time herein are novel proteins that are expressed in, inter alia, human cell lines and human brain, pituitary, cerebellum, spinal cord, lymph node, testis, adrenal gland, uterus, mammary gland, rectum, hypothalamus, ovary, fetal kidney, fetal lung, aorta, 6-, 9-, and 12-week old embryos, adenocarcinoma, osteosarcoma, embryonic carcinoma, and normal umbilical vein cells.
  • the described sequences were compiled from sequences available in GENBANK, and cDNAs generated from human adult and fetal brain, pituitary, hypothalamus, testis, and fetus mRNAs (Edge Biosystems, Gaithersburg, Md.) that were identified using primers generated from human genomic DNA.
  • the present invention encompasses the nucleotides presented in the Sequence Listing, host cells expressing such nucleotides, the expression products of such nucleotides, and: (a) nucleotides that encode mammalian homologs of the described polynucleotides, including the specifically described NHPs, and the NHP products; (b) nucleotides that encode one or more portions of a NHP that correspond to any functional domain(s), and the polypeptide products specified by such nucleotide sequences, including, but not limited to, the novel regions of any active domain(s); (c) isolated nucleotides that encode mutant versions, engineered or naturally occurring, of the described NHPs, in which all or a part of at least one domain is deleted or altered, and the polypeptide products specified by such nucleotide sequences, including, but not limited to, soluble proteins and peptides; (d) nucleotides that encode chimeric fusion proteins containing all or a portion of a
  • the present invention includes the human DNA sequences presented in the Sequence Listing (and vectors comprising the same), and additionally contemplates any nucleotide sequence encoding a contiguous NHP open reading frame (ORF) that hybridizes to a complement of a DNA sequence presented in the Sequence Listing under highly stringent conditions, e.g., hybridization to filter-bound DNA in 0.5 M NaHPO 4 , 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65° C., and washing in 0.1x SSC/0.1% SDS at 68° C. (Ausubel et al., eds., 1989, Current Protocols in Molecular Biology, Vol.
  • ORF NHP open reading frame
  • NHP NHP polynucleotide sequences.
  • mutant NHPs whether naturally occurring or engineered (by site directed mutagenesis, gene shuffling, directed evolution as described in, for example, U.S. Pat. Nos. 5,837,458 or 5,723,323, both of which are herein incorporated by reference).
  • the invention also includes degenerate nucleic acid variants of the disclosed NHP polynucleotide sequences.
  • polynucleotides encoding NHP ORFs, or their functional equivalents encoded by polynucleotide sequences that are about 99, 95, 90, or about 85 percent similar to corresponding regions of SEQ ID NOS:1, 3 or 5 (as measured by BLAST sequence comparison analysis using, for example, the GCG sequence analysis package, as described herein, using default parameters).
  • the invention also includes nucleic acid molecules, preferably DNA molecules, that hybridize to, and are therefore the complements of, the described NHP-encoding polynucleotides.
  • Such hybridization conditions can be highly stringent or less highly stringent, as described herein.
  • the nucleic acid molecules are deoxyoligonucleotides (“DNA oligos”)
  • DNA oligos” such molecules are generally about 16 to about 100 bases long, or about 20 to about 80 bases long, or about 34 to about 45 bases long, or any variation or combination of sizes represented therein that incorporate a contiguous region of sequence first disclosed in the Sequence Listing.
  • Such oligonucleotides can be used in conjunction with the polymerase chain reaction (PCR) to screen libraries, isolate clones, and prepare cloning and sequencing templates, etc.
  • PCR polymerase chain reaction
  • NHP oligonucleotides can be used as hybridization probes for screening libraries, and assessing gene expression patterns (particularly using a microarray or high-throughput “chip” format).
  • a series of NHP oligonucleotide sequences, or the complements thereof, can be used to represent all or a portion of the described NHP sequences.
  • An oligonucleotide or polynucleotide sequence first disclosed in at least a portion of one or more of the sequences of SEQ ID NOS:1-6 can be used as a hybridization probe, in conjunction with a solid support matrix/substrate (resins, beads, membranes, plastics, polymers, metal or metallized substrates, crystalline or polycrystalline substrates, etc.).
  • a solid support matrix/substrate resins, beads, membranes, plastics, polymers, metal or metallized substrates, crystalline or polycrystalline substrates, etc.
  • spatially addressable arrays i.e., gene chips, microtiter plates, etc.
  • oligonucleotides and polynucleotides or corresponding oligopeptides and polypeptides
  • at least one of the biopolymers present on the spatially addressable array comprises an oligonucleotide or polynucleotide sequence first disclosed in at least one of the sequences of SEQ ID NOS:1-6, or an amino acid sequence encoded thereby.
  • Addressable arrays comprising sequences first disclosed in SEQ ID NOS:1-6 can be used to identify and characterize the temporal and tissue specific expression of a gene. These addressable arrays incorporate oligonucleotide sequences of sufficient length to confer the required specificity, yet be within the limitations of the production technology. The length of these probes is usually within a range of between about 8 to about 2000 nucleotides. Preferably the probes consist of 60 nucleotides, and more preferably 25 nucleotides, from the sequences first disclosed in SEQ ID NOS:1-6.
  • a series of NHP oligonucleotide sequences can be used in chip format to represent all or a portion of the described sequences.
  • the oligonucleotides typically between about 16 to about 40 (or any whole number within the stated range) nucleotides in length, can partially overlap each other, and/or the sequence may be represented using oligonucleotides that do not overlap.
  • the described polynucleotide sequences shall typically comprise at least about two or three distinct oligonucleotide sequences of at least about 8 nucleotides in length that are each first disclosed in the described Sequence Listing.
  • Such oligonucleotide sequences can begin at any nucleotide present within a sequence in the Sequence Listing, and proceed in either a sense (5′-to-3′) orientation vis-a-vis the described sequence or in an antisense orientation.
  • Microarray-based analysis allows the discovery of broad patterns of genetic activity, providing new understanding of gene functions, and generating novel and unexpected insight into transcriptional processes and biological mechanisms.
  • the use of addressable arrays comprising sequences first disclosed in SEQ ID NOS:1-6 provides detailed information about transcriptional changes involved in a specific pathway, potentially leading to the identification of novel components, or gene functions that manifest themselves as novel phenotypes.
  • Probes consisting of sequences first disclosed in SEQ ID NOS:1-6 can also be used in the identification, selection, and validation of novel molecular targets for drug discovery.
  • the use of these unique sequences permits the direct confirmation of drug targets, and recognition of drug dependent changes in gene expression that are modulated through pathways distinct from the intended target of the drug. These unique sequences therefore also have utility in defining and monitoring both drug action and toxicity.
  • sequences first disclosed in SEQ ID NOS:1-6 can be utilized in microarrays, or other assay formats, to screen collections of genetic material from patients who have a particular medical condition. These investigations can also be carried out using the sequences first disclosed in SEQ ID NOS:1-6 in silico, and by comparing previously collected genetic databases and the disclosed sequences using computer software known to those in the art.
  • sequences first disclosed in SEQ ID NOS:1-6 can be used to identify mutations associated with a particular disease, and also in diagnostic or prognostic assays.
  • sequences have been specifically described using nucleotide sequence, it should be appreciated that each of the sequences can uniquely be described using any of a wide variety of additional structural attributes, or combinations thereof.
  • a given sequence can be described by the net composition of the nucleotides present within a given region of the sequence, in conjunction with the presence of one or more specific oligonucleotide sequence(s) first disclosed in SEQ ID NOS:1-6.
  • a restriction map specifying the relative positions of restriction endonuclease digestion sites, or various palindromic or other specific oligonucleotide sequences, can be used to structurally describe a given sequence.
  • restriction maps which are typically generated by widely available computer programs (e.g., the University of Wisconsin GCG sequence analysis package, SEQUENCHER 3.0, Gene Codes Corp., Ann Arbor, Mich., etc.), can optionally be used in conjunction with one or more discrete nucleotide sequence(s) present in the sequence that can be described by the relative position of the sequence relative to one or more additional sequence(s) or one or more restriction sites present in the disclosed sequence.
  • highly stringent conditions may refer, e.g., to washing in 6x SSC/0.05% sodium pyrophosphate at 37° C. (for 14-base oligos), 48° C. (for 17-base oligos), 55° C. (for 20-base oligos), and 60° C. (for 23-base oligos).
  • These nucleic acid molecules may encode or act as NHP antisense molecules, useful, for example, in NHP gene regulation and/or as antisense primers in amplification reactions of NHP nucleic acid sequences.
  • NHP gene regulation such techniques can be used to regulate biological functions.
  • sequences can be used as part of ribozyme and/or triple helix sequences that are also useful for NHP gene regulation.
  • Inhibitory antisense or double stranded oligonucleotides can additionally comprise at least one modified base moiety that is selected from the group including, but not limited to, 5-fluorouracil, 5,-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-
  • the antisense oligonucleotide can also comprise at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the antisense oligonucleotide will comprise at least one modified phosphate backbone selected from the group including, but not limited to, a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof.
  • the antisense oligonucleotide is an ⁇ -anomeric oligonucleotide.
  • An ⁇ -anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641).
  • the oligonucleotide is a 2′-0-methylribonucleotide (Inoue et al., 1987, Nucl. Acids Res.
  • RNA-DNA analogue a chimeric RNA-DNA analogue
  • double stranded RNA can be used to disrupt the expression and function of a targeted NHP.
  • Oligonucleotides of the invention can be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.).
  • an automated DNA synthesizer such as are commercially available from Biosearch, Applied Biosystems, etc.
  • phosphorothioate oligonucleotides can be synthesized by the method of Stein et al. (1988, Nucl. Acids Res. 16:3209), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., 1988, Proc. Natl. Acad. Sci. USA 85:7448-7451), etc.
  • Low stringency conditions are well-known to those of skill in the art, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived. For guidance regarding such conditions see, for example, Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (and periodic updates thereof), and Ausubel et al., 1989, supra.
  • NHP nucleotide probes can be used to screen a human genomic library using appropriately stringent conditions or by PCR.
  • the identification and characterization of human genomic clones is helpful for identifying polymorphisms (including, but not limited to, nucleotide repeats, microsatellite alleles, single nucleotide polymorphisms, or coding single nucleotide polymorphisms), determining the genomic structure of a given locus/allele, and designing diagnostic tests.
  • sequences derived from regions adjacent to the intron/exon boundaries of the human gene can be used to design primers for use in amplification assays to detect mutations within the exons, introns, splice sites (e.g., splice acceptor and/or donor sites), etc., that can be used in diagnostics and pharmacogenomics.
  • splice sites e.g., splice acceptor and/or donor sites
  • the present sequences can be used in restriction fragment length polymorphism (RFLP) analysis to identify specific individuals.
  • RFLP restriction fragment length polymorphism
  • an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification (as generally described in U.S. Pat. No. 5,272,057, incorporated herein by reference).
  • the sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another “identification marker” (i.e., another DNA sequence that is unique to a particular individual).
  • another “identification marker” i.e., another DNA sequence that is unique to a particular individual.
  • Actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments.
  • a NHP homolog can be isolated from nucleic acid from an organism of interest by performing PCR using two degenerate or “wobble” oligonucleotide primer pools designed on the basis of amino acid sequences within the NHP products disclosed herein.
  • the template for the reaction may be total RNA, mRNA, genomic DNA and/or cDNA obtained by reverse transcription of mRNA prepared from, for example, human or non-human cell lines or tissue known to express, or suspected of expressing, an allele of a NHP gene.
  • the PCR product can be subcloned and sequenced to ensure that the amplified sequences represent the sequence of the desired NHP gene.
  • the PCR fragment can then be used to isolate a full length cDNA clone by a variety of methods.
  • the amplified fragment can be labeled and used to screen a cDNA library, such as a bacteriophage cDNA library.
  • the labeled fragment can be used to isolate genomic clones via the screening of a genomic library.
  • RNA can be isolated, following standard procedures, from an appropriate cellular or tissue source (i.e., one known to express, or suspected of expressing, a NHP gene).
  • a reverse transcription (RT) reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5′ end of the amplified fragment for the priming of first strand synthesis.
  • the resulting RNA/DNA hybrid may then be “tailed” using a standard terminal transferase reaction, the hybrid may be digested with RNase H, and second strand synthesis may then be primed with a complementary primer.
  • cDNA sequences upstream of the amplified fragment can be isolated.
  • a cDNA encoding a mutant NHP sequence can be isolated, for example, by using PCR.
  • the first cDNA strand may be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue known to express, or suspected of expressing, a NHP, in an individual putatively carrying a mutant NHP allele, and by extending the new strand with reverse transcriptase.
  • the second strand of the cDNA is then synthesized using an oligonucleotide that hybridizes specifically to the 5′ end of the normal sequence.
  • the product is then amplified via PCR, optionally cloned into a suitable vector, and subjected to DNA sequence analysis through methods well-known to those of skill in the art.
  • DNA sequence analysis By comparing the DNA sequence of the mutant NHP allele to that of a corresponding normal NHP allele, the mutation(s) responsible for the loss or alteration of function of the mutant NHP gene product can be ascertained.
  • a genomic library can be constructed using DNA obtained from an individual suspected of carrying, or known to carry, a mutant NHP allele (e.g., a person manifesting a NHP-associated phenotype such as, for example, behavioral disorders, immune disorders, obesity, high blood pressure, etc.), or a cDNA library can be constructed using RNA from a tissue known to express, or suspected of expressing, a mutant NHP allele.
  • a normal NHP gene, or any suitable fragment thereof, can then be labeled and used as a probe to identify the corresponding mutant NHP allele in such libraries.
  • Clones containing mutant NHP sequences can then be purified and subjected to sequence analysis according to methods well-known to those skilled in the art.
  • an expression library can be constructed utilizing cDNA synthesized from, for example, RNA isolated from a tissue known to express, or suspected of expressing, a mutant NHP allele in an individual suspected of carrying, or known to carry, such a mutant allele.
  • gene products made by the putatively mutant tissue may be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against a normal NHP product, as described below (for screening techniques, see, for example, Harlow and Lane, eds., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Press, Cold Spring Harbor, N.Y.).
  • screening can be accomplished by screening with labeled NHP fusion proteins, such as, for example, alkaline phosphatase-NHP or NHP-alkaline phosphatase fusion proteins.
  • labeled NHP fusion proteins such as, for example, alkaline phosphatase-NHP or NHP-alkaline phosphatase fusion proteins.
  • polyclonal antibodies to a NHP are likely to cross-react with a corresponding mutant NHP expression product.
  • Library clones detected via their reaction with such labeled antibodies can be purified and subjected to sequence analysis according to methods well-known in the art.
  • the invention also encompasses: (a) DNA vectors that contain any of the foregoing NHP coding sequences and/or their complements (i.e., antisense); (b) DNA expression vectors that contain any of the foregoing NHP coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences (for example, baculovirus as described in U.S. Pat. No.
  • regulatory elements include, but are not limited to, inducible and non-inducible promoters, enhancers, operators, and other elements known to those skilled in the art that drive and regulate expression.
  • Such regulatory elements include, but are not limited to, the cytomegalovirus (hCMV) immediate early gene, regulatable, viral elements (particularly retroviral LTR promoters), the early or late promoters of SV40 or adenovirus, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage lambda, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase (PGK), the promoters of acid phosphatase, and the promoters of the yeast ⁇ -mating factors.
  • hCMV cytomegalovirus
  • regulatable, viral elements particularly retroviral LTR promoters
  • the early or late promoters of SV40 or adenovirus the lac system
  • the trp system the TAC system
  • TRC system the major operator and promoter regions of phage lambda
  • the control regions of fd coat protein the promoter for 3-phosphoglycer
  • NHP peptides or polypeptides are thought to be cytoplasmic or nuclear proteins (although processed forms or fragments can be secreted or membrane associated)
  • expression systems can be engineered that produce soluble derivatives of a NHP (corresponding to a NHP extracellular and/or intracellular domains, or truncated polypeptides lacking one or more hydrophobic domains) and/or NHP fusion protein products (especially NHP-Ig fusion proteins, i.e., fusions of a NHP domain to an IgFc), NHP antibodies, and anti-idiotypic antibodies (including Fab fragments) that can be used in therapeutic applications.
  • the above expression systems are engineered to allow the desired peptide or polypeptide to be recovered from the culture media.
  • the present invention also encompasses antibodies and anti-idiotypic antibodies (including Fab fragments), antagonists and agonists of a NHP, as well as compounds or nucleotide constructs that inhibit expression of a NHP sequence (transcription factor inhibitors, antisense and ribozyme molecules, or open reading frame sequence or regulatory sequence replacement constructs), or promote the expression of a NHP (e.g., expression constructs in which NHP coding sequences are operatively associated with expression control elements such as promoters, promoter/enhancers, etc.).
  • a NHP sequence transcription factor inhibitors, antisense and ribozyme molecules, or open reading frame sequence or regulatory sequence replacement constructs
  • promote the expression of a NHP e.g., expression constructs in which NHP coding sequences are operatively associated with expression control elements such as promoters, promoter/enhancers, etc.
  • the NHPs or NHP peptides, NHP fusion proteins, NHP nucleotide sequences, antibodies, antagonists and agonists can be useful for the detection of mutant NHPs, or inappropriately expressed NHPs, for the diagnosis of disease.
  • the NHP proteins or peptides, NHP fusion proteins, NHP nucleotide sequences, host cell expression systems, antibodies, antagonists, agonists and genetically engineered cells and animals can be used for screening for drugs (or high throughput screening of combinatorial libraries) effective in the treatment of the symptomatic or phenotypic manifestations of perturbing the normal function of a NHP in the body.
  • the use of engineered host cells and/or animals can offer an advantage in that such systems allow not only for the identification of compounds that bind to the endogenous receptor/ligand of a NHP, but can also identify compounds that trigger NHP-mediated activities or pathways.
  • NHP products can be used as therapeutics.
  • soluble derivatives such as NHP peptides/domains corresponding to NHPs, NHP fusion protein products (especially NHP-Ig fusion proteins, i.e., fusions of a NHP, or a domain of a NHP, to an IgFc), NHP antibodies and anti-idiotypic antibodies (including Fab fragments), antagonists or agonists (including compounds that modulate or act on downstream targets in a NHP-mediated pathway) can be used to directly treat diseases or disorders.
  • NHP fusion protein products especially NHP-Ig fusion proteins, i.e., fusions of a NHP, or a domain of a NHP, to an IgFc
  • NHP antibodies and anti-idiotypic antibodies including Fab fragments
  • antagonists or agonists including compounds that modulate or act on downstream targets in a NHP-mediated pathway
  • nucleotide constructs encoding such NHP products can be used to genetically engineer host cells to express such products in vivo; these genetically engineered cells function as “bioreactors” in the body delivering a continuous supply of a NHP, a NHP peptide, or a NHP fusion protein to the body.
  • Nucleotide constructs encoding functional NHPs, mutant NHPs, as well as antisense and ribozyme molecules, can also be used in “gene therapy” approaches for the modulation of NHP expression.
  • the invention also encompasses pharmaceutical formulations and methods for treating biological disorders.
  • the described NHPs can be expressed in a moderate range of human tissues.
  • the described NHPs also share significant similarity to a range of additional kinase families, including kinases associated with signal transduction, from a variety of phyla and species.
  • the genomic locus encoding the described NHPs is apparently encoded on human chromosome 19 (see GENBANK accession no. AC020922). As such, the described sequences are useful, inter alia, for mapping the coding regions of the human genome, and particularly chromosome 19.
  • NHP gene products can also be expressed in transgenic animals.
  • Animals of any species including, but not limited to, worms, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, birds, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, may be used to generate NHP transgenic animals.
  • Any technique known in the art may be used to introduce a NHP transgene into animals to produce the founder lines of transgenic animals.
  • Such techniques include, but are not limited to, pronuclear microinjection (Hoppe and Wagner, 1989, U.S. Pat. No. 4,873,191); retrovirus-mediated gene transfer into germ lines (Van der Putten et al., 1985, Proc. Natl. Acad. Sci. USA 82:6148-6152); gene targeting in embryonic stem cells (Thompson et al., 1989, Cell 56:313-321); electroporation of embryos (Lo, 1983, Mol Cell. Biol.
  • the present invention provides for transgenic animals that carry a NHP transgene in all their cells, as well as animals that carry a transgene in some, but not all their cells, i.e., mosaic animals or somatic cell transgenic animals.
  • a transgene may be integrated as a single transgene, or in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
  • a transgene may also be selectively introduced into and activated in a particular cell-type by following, for example, the teaching of Lasko et al., 1992, Proc. Natl. Acad. Sci. USA 89:6232-6236. The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell-type of interest, and will be apparent to those of skill in the art.
  • a NHP transgene be integrated into the chromosomal site of the endogenous NHP gene
  • gene targeting is preferred.
  • vectors containing some nucleotide sequences homologous to the endogenous NHP gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous NHP gene (i.e., “knockout” animals).
  • the transgene can also be selectively introduced into a particular cell-type, thus inactivating the endogenous NHP gene in only that cell-type, by following, for example, the teaching of Gu et al., 1994, Science 265:103-106.
  • the regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell-type of interest, and will be apparent to those of skill in the art.
  • the expression of the recombinant NHP sequence may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques that include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and RT-PCR. Samples of NHP gene-expressing tissue may also be evaluated immunocytochemically using antibodies specific for the NHP transgene product.
  • the present invention also provides for “knock-in” animals.
  • Knock-in animals are those in which a polynucleotide sequence (i.e., a gene or a cDNA) that the animal does not naturally have in its genome is inserted in such a way that it is expressed. Examples include, but are not limited to, a human gene or cDNA used to replace its murine ortholog in the mouse, a murine cDNA used to replace the murine gene in the mouse, and a human gene or cDNA or murine cDNA that is tagged with a reporter construct used to replace the murine ortholog or gene in the mouse. Such replacements can occur at the locus of the murine ortholog or gene, or at another specific site.
  • Such knock-in animals are useful for the in vivo study, testing and validation of, intra alia, human drug targets, as well as for compounds that are directed at the same, and therapeutic proteins.
  • NHPs, NHP polypeptides, NHP peptide fragments, mutated, truncated, or deleted forms of the NHPs, and/or NHP fusion proteins can be prepared for a variety of uses. These uses include, but are not limited to, the generation of antibodies, as reagents in diagnostic assays, for the identification of other cellular gene products related to a NHP, and as reagents in assays for screening for compounds that can be used as pharmaceutical reagents useful in the therapeutic treatment of mental, biological, or medical disorders and diseases. Given the similarity information and expression data, the described NHPs can be targeted (by drugs, oligonucleotides, antibodies, etc.) in order to treat disease, or to therapeutically augment the efficacy of therapeutic agents.
  • the Sequence Listing discloses the amino acid sequences encoded by the described NHP-encoding polynucleotides.
  • the NHPs display initiator methionines that are present in DNA sequence contexts consistent with eucaryotic translation initiation sites.
  • the NHPs do not display consensus signal sequences, which indicates that they may be cytoplasmic or possibly nuclear proteins, although they may also be secreted or membrane associated (given the presence of several hydrophobic domains).
  • NHP amino acid sequences of the invention include the amino acid sequences presented in the Sequence Listing, as well as analogues and derivatives thereof. Further, corresponding NHP homologues from other species are encompassed by the invention.
  • any NHP protein encoded by the NHP nucleotide sequences described herein are within the scope of the invention, as are any novel polynucleotide sequences encoding all or any novel portion of an amino acid sequence presented in the Sequence Listing.
  • the degenerate nature of the genetic code is well-known, and, accordingly, each amino acid presented in the Sequence Listing is generically representative of the well-known nucleic acid “triplet” codon, or in many cases codons, that can encode the amino acid.
  • amino acid sequences presented in the Sequence Listing when taken together with the genetic code (see, for example, Table 4-1 at page 109 of “Molecular Cell Biology”, 1986, J. Darnell et al., eds., Scientific American Books, New York, N.Y., herein incorporated by reference), are generically representative of all the various permutations and combinations of nucleic acid sequences that can encode such amino acid sequences.
  • the invention also encompasses proteins that are functionally equivalent to the NHPs encoded by the presently described nucleotide sequences, as judged by any of a number of criteria, including, but not limited to, the ability to bind and modify a NHP substrate, or the ability to effect an identical or complementary downstream pathway, or a change in cellular metabolism (e.g., proteolytic activity, ion flux, phosphorylation, etc.).
  • Such functionally equivalent NHP proteins include, but are not limited to, additions or substitutions of amino acid residues within the amino acid sequence encoded by the NHP nucleotide sequences described herein, but that result in a silent change, thus producing a functionally equivalent expression product.
  • Nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine
  • polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine
  • positively charged (basic) amino acids include arginine, lysine, and histidine
  • negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • a variety of host-expression vector systems can be used to express the NHP nucleotide sequences of the invention. Where the NHP peptide or polypeptide can exist, or has been engineered to exist, as a soluble or secreted molecule, the soluble NHP peptide or polypeptide can be recovered from the culture media.
  • Such expression systems also encompass engineered host cells that express a NHP, or functional equivalent, in situ. Purification or enrichment of a NHP from such expression systems can be accomplished using appropriate detergents and lipid micelles and methods well-known to those skilled in the art. However, such engineered host cells themselves may be used in situations where it is important not only to retain the structural and functional characteristics of a NHP, but to assess biological activity, e.g., in certain drug screening assays.
  • the expression systems that may be used for purposes of the invention include, but are not limited to, microorganisms such as bacteria (e.g., E. coli, B. subtilis ) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing NHP nucleotide sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing NHP nucleotide sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing NHP nucleotide sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing NHP nucleotide sequences; or mammalian cell systems (e.g.
  • a number of expression vectors may be advantageously selected depending upon the use intended for the NHP product being expressed. For example, when a large quantity of such a protein is to be produced for the generation of pharmaceutical compositions of or containing a NHP, or for raising antibodies to a NHP, vectors that direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • vectors include, but are not limited to, the E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J.
  • a NHP coding sequence may be ligated individually into the vector in-frame with the lacZ coding region so that a fusion protein is produced; pIN vectors (Inouye and Inouye, 1985, Nucleic Acids Res. 13:3101-3109; Van Heeke and Schuster, 1989, J. Biol. Chem. 264:5503-5509); and the like.
  • PGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target expression product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign polynucleotide sequences.
  • the virus grows in Spodoptera frugiperda cells.
  • a NHP coding sequence can be cloned individually into a non-essential region (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • Successful insertion of a NHP coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene).
  • the NHP nucleotide sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric sequence may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • an adenovirus transcription/translation control complex e.g., the late promoter and tripartite leader sequence.
  • This chimeric sequence may then be inserted in the adenovirus genome by in vitro or in vivo recombination.
  • Insertion in a non-essential region of the viral genome will result in a recombinant virus that is viable and capable of expressing a NHP product in infected hosts (e.g., see Logan and Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:3655-3659).
  • Specific initiation signals may also be required for efficient translation of inserted NHP nucleotide sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire NHP gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed.
  • exogenous translational control signals including, perhaps, the ATG initiation codon
  • the initiation codon should be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert.
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., 1987, Methods in Enzymol. 153:516-544).
  • a host cell strain may be chosen that modulates the expression of the inserted sequences, or modifies and processes the expression product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and expression products. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed.
  • eukaryotic host cells that possess the cellular machinery for the desired processing of the primary transcript, glycosylation, and phosphorylation of the expression product may be used.
  • mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, and in particular, human cell lines.
  • cell lines that stably express the NHP sequences described herein can be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci, which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines that express the NHP product.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the NHP product.
  • a number of selection systems may be used, including, but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska and Szybalski, 1962, Proc. Natl. Acad. Sci. USA 48:2026), and adenine, phosphoribosyltransferase (Lowy et al., 1980, Cell 22:817) genes, which can be employed in tk ⁇ , hgprt ⁇ or aprt ⁇ cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., 1980, Proc. Natl. Acad. Sci. USA 77:3567; O'Hare et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan and Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., 1981, J. Mol. Biol. 150:1); and hygro, which confers resistance to hygromycin (Santerre et al., 1984, Gene 30:147).
  • any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed.
  • Another exemplary system allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972-8976).
  • the sequence of interest is subcloned into a vaccinia recombination plasmid such that the sequence's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni 2+ ⁇ nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • fusion proteins that direct a NHP to a target organ and/or facilitate transport across the membrane into the cytosol.
  • Conjugation of NHPs to antibody molecules or their Fab fragments could be used to target cells bearing a particular epitope. Attaching an appropriate signal sequence to a NHP would also transport a NHP to a desired location within the cell.
  • targeting of a NHP or its nucleic acid sequence might be achieved using liposome or lipid complex based delivery systems.
  • liposome or lipid complex based delivery systems are described in “Liposomes: A Practical Approach”, New, R.R.C., ed., Oxford University Press, N.Y., and in U.S. Pat. Nos.
  • novel protein constructs engineered in such a way that they facilitate transport of NHPs to a target site or desired organ, where they cross the cell membrane and/or the nucleus where the NHPs can exert their functional activity.
  • This goal may be achieved by coupling of a NHP to a cytokine or other ligand that provides targeting specificity, and/or to a protein transducing domain (see generally U.S. Provisional Patent Application Ser. Nos. 60/111,701 and 60/056,713, both of which are herein incorporated by reference, for examples of such transducing sequences), to facilitate passage across cellular membranes, and can optionally be engineered to include nuclear localization signals.
  • oligopeptides that are modeled on an amino acid sequence first described in the Sequence Listing.
  • Such NHP oligopeptides are generally between about 10 to about 100 amino acids long, or between about 16 to about 80 amino acids long, or between about 20 to about 35 amino acids long, or any variation or combination of sizes represented therein that incorporate a contiguous region of sequence first disclosed in the Sequence Listing.
  • Such NHP oligopeptides can be of any length disclosed within the above ranges and can initiate at any amino acid position represented in the Sequence Listing.
  • the invention also contemplates “substantially isolated” or “substantially pure” proteins or polypeptides.
  • a “substantially isolated” or “substantially pure” protein or polypeptide is meant a protein or polypeptide that has been separated from at least some of those components which naturally accompany it.
  • the protein or polypeptide is substantially isolated or pure when it is at least 60%, by weight, free from the proteins and other naturally-occurring organic molecules with which it is naturally associated in vivo.
  • the purity of the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight.
  • a substantially isolated or pure protein or polypeptide may be obtained, for example, by extraction from a natural source, by expression of a recombinant nucleic acid encoding the protein or polypeptide, or by chemically synthesizing the protein or polypeptide.
  • Purity can be measured by any appropriate method, e.g., column chromatography such as immunoaffinity chromatography using an antibody specific for the protein or polypeptide, polyacrylamide gel electrophoresis, or HPLC analysis.
  • a protein or polypeptide is substantially free of naturally associated components when it is separated from at least some of those contaminants which accompany it in its natural state.
  • a polypeptide which is chemically synthesized or produced in a cellular system different from the cell from which it naturally originates will be, by definition, substantially free from its naturally associated components.
  • substantially isolated or pure proteins or polypeptides include eukaryotic proteins synthesized in E. coli, other prokaryotes, or any other organism in which they do not naturally occur.
  • Antibodies that specifically recognize one or more epitopes of a NHP, epitopes of conserved variants of a NHP, or peptide fragments of a NHP are also encompassed by the invention.
  • Such antibodies include, but are not limited to, polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′) 2 fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • the antibodies of the invention can be used, for example, in the detection of a NHP in a biological sample and may, therefore, be utilized as part of a diagnostic or prognostic technique whereby patients may be tested for abnormal amounts of a NHP.
  • Such antibodies may also be utilized in conjunction with, for example, compound screening schemes for the evaluation of the effect of test compounds on expression and/or activity of a NHP expression product.
  • such antibodies can be used in conjunction with gene therapy to, for example, evaluate normal and/or engineered NHP-expressing cells prior to their introduction into a patient.
  • Such antibodies may additionally be used in methods for the inhibition of abnormal NHP activity.
  • Such antibodies may be utilized as a part of treatment methods.
  • various host animals may be immunized by injection with a NHP, a NHP peptide (e.g., one corresponding to a functional domain of a NHP), truncated NHP polypeptides (NHP in which one or more domains have been deleted), functional equivalents of a NHP, or mutated variants of a NHP.
  • a NHP peptide e.g., one corresponding to a functional domain of a NHP
  • NHP truncated NHP polypeptides
  • Such host animals may include, but are not limited to, pigs, rabbits, mice, goats, and rats, to name but a few.
  • adjuvants may be used to increase the immunological response, depending on the host species, including, but not limited to, Freund's adjuvant (complete and incomplete), mineral salts such as aluminum hydroxide or aluminum phosphate, chitosan, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.
  • BCG Bacille Calmette-Guerin
  • Corynebacterium parvum bacille Calmette-Guerin
  • the immune response could be enhanced by combination and/or coupling with molecules such as keyhole limpet hemocyanin, tetanus toxoid, diphtheria toxoid, ovalbumin, cholera toxin, or fragments thereof.
  • molecules such as keyhole limpet hemocyanin, tetanus toxoid, diphtheria toxoid, ovalbumin, cholera toxin, or fragments thereof.
  • Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of the immunized animals.
  • Monoclonal antibodies which are homogeneous populations of antibodies to a particular antigen, can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Pat. No. 4,376,110), the human B-cell hybridoma technique (Kosbor et al., 1983, Immunology Today 4:72; Cole et al., 1983, Proc. Natl. Acad. Sci. USA 80:2026-2030), and the EBV-hybridoma technique (Cole. et al., 1985, Monoclonal Antibodies And Cancer Therapy, Alan R.
  • Such antibodies may be of any immunoglobulin class, including IgG, IgM, IgE, IgA, and IgD, and any subclass thereof.
  • the hybridomas producing the mAbs of this invention may be cultivated in vitro or in vivo. Production of high titers of mAbs in vivo makes this the presently preferred method of production.
  • chimeric antibodies In addition, techniques developed for the production of “chimeric antibodies” (Morrison et al., 1984, Proc. Natl. Acad. Sci. USA 81:6851-6855; Neuberger. et al., 1984, Nature, 312:604-608; Takeda et al., 1985, Nature, 314:452-454), by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity, can be used.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region. Such technologies are described in U.S.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Antibody fragments that recognize specific epitopes may be generated by known techniques.
  • fragments include, but are not limited to: F(ab′) 2 fragments, which can be produced by pepsin digestion of an antibody molecule; and Fab fragments, which can be generated by reducing the disulfide bridges of F(ab′) 2 fragments.
  • Fab expression libraries may be constructed (Huse et al., 1989, Science, 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
  • Antibodies to a NHP can, in turn, be utilized to generate anti-idiotype antibodies that “mimic” a given NHP, using techniques well-known to those skilled in the art (see, e.g., Greenspan and Bona, 1993, FASEB J. 7:437-444; and Nissinoff, 1991, J. Immunol. 147:2429-2438).
  • antibodies that bind to a NHP domain and competitively inhibit the binding of a NHP to its cognate receptor/ligand can be used to generate anti-idiotypes that “mimic” the NHP and, therefore, bind, activate, or neutralize a NHP, NHP receptor, or NHP ligand.
  • Such anti-idiotypic antibodies or Fab fragments of such anti-idiotypes can be used in therapeutic regimens involving a NHP-mediated pathway.
  • the presently described knock-out mice have a unique utility, as they can be advantageously applied to the generation of antibodies against the disclosed mammalian NHPs (i.e., a NHP will be immunogenic in NHP knock-out animals).

Abstract

Novel human polynucleotide and polypeptide sequences are disclosed that can be used in therapeutic, diagnostic, and pharmacogenomic applications.

Description

  • The present application claims the benefit of U.S. Provisional Application No. 60/282,036, which was filed on Apr. 6, 2001, and is herein incorporated by reference in its entirety.[0001]
  • INTRODUCTION
  • The present invention relates to the discovery, identification, and characterization of novel human polynucleotides encoding proteins sharing sequence similarity with animal kinases. The invention encompasses the described polynucleotides, host cell expression systems, the encoded proteins, fusion proteins, polypeptides and peptides, antibodies to the encoded proteins and peptides, and genetically engineered animals that either lack or overexpress the disclosed polynucleotides, antagonists and agonists of the proteins, and other compounds that modulate the expression or activity of the proteins encoded by the disclosed polynucleotides, which can be used for diagnosis, drug screening, clinical trial monitoring, the treatment of diseases and disorders, and cosmetic or nutriceutical applications. [0002]
  • BACKGROUND OF THE INVENTION
  • Kinases mediate the phosphorylation of a wide variety of proteins and compounds in the cell. Along with phosphatases, kinases are involved in a range of regulatory pathways. Given the physiological importance of kinases, they have been subject to intense scrutiny and are proven drug targets. [0003]
  • SUMMARY OF THE INVENTION
  • The present invention relates to the discovery, identification, and characterization of nucleotides that encode novel human proteins and the corresponding amino acid sequences of these proteins. The novel human proteins (NHPs), described for the first time herein, share structural similarity with animal kinases, including, but not limited to, serine-threonine kinases, and carbon catabolite depressing kinases. Accordingly, the described NHPs encode novel kinases having homologues and orthologs across a range of phyla and species. [0004]
  • The novel human polynucleotides described herein encode open reading frames (ORFs) encoding proteins of 778, 762, and 703 amino acids in length (see respectively SEQ ID NOS:2, 4, and 6). [0005]
  • The invention also encompasses agonists and antagonists of the described NHPs, including small molecules, large molecules, mutant NHPs, or portions thereof, that compete with native NHPs, peptides, and antibodies, as well as nucleotide sequences that can be used to inhibit the expression of the described NHPs (e.g., antisense and ribozyme molecules, and open reading frame or regulatory sequence replacement constructs) or to enhance the expression of the described NHPs (e.g., expression constructs that place the described polynucleotide under the control of a strong promoter system), and transgenic animals that express a NHP sequence, or “knock-outs” (which can be conditional) that do not express a functional NHP. Knock-out mice can be produced in several ways, one of which involves the use of mouse embryonic stem cell (“ES cell”) lines that contain gene trap mutations in a murine homolog of at least one of the described NHPs. When the unique NHP sequences described in SEQ ID NOS:1-6 are “knocked-out” they provide a method of identifying phenotypic expression of the particular gene, as well as a method of assigning function to previously unknown genes. In addition, animals in which the unique NHP sequences described in SEQ ID NOS:1-6 are “knocked-out” provide a unique source in which to elicit antibodies to homologous and orthologous proteins, which would have been previously viewed by the immune system as “self” and therefore would have failed to elicit significant antibody responses. To these ends, gene trapped knockout ES cells have been generated in murine homologs of the described NHPs. [0006]
  • Additionally, the unique NHP sequences described in SEQ ID NOS:1-6 are useful for the identification of protein coding sequences, and mapping a unique gene to a particular chromosome. These sequences identify biologically verified exon splice junctions, as opposed to splice junctions that may have been bioinformatically predicted from genomic sequence alone. The sequences of the present invention are also useful as additional DNA markers for restriction fragment length polymorphism (RFLP) analysis, and in forensic biology. [0007]
  • Further, the present invention also relates to processes for identifying compounds that modulate, i.e., act as agonists or antagonists of, NHP expression and/or NHP activity that utilize purified preparations of the described NHPs and/or NHP products, or cells expressing the same. Such compounds can be used as therapeutic agents for the treatment of any of a wide variety of symptoms associated with biological disorders or imbalances. [0008]
  • DESCRIPTION OF THE SEQUENCE LISTING AND FIGURES
  • The Sequence Listing provides the sequence of the novel human ORFs encoding the described novel human kinase proteins.[0009]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The NHPs described for the first time herein are novel proteins that are expressed in, inter alia, human cell lines and human brain, pituitary, cerebellum, spinal cord, lymph node, testis, adrenal gland, uterus, mammary gland, rectum, hypothalamus, ovary, fetal kidney, fetal lung, aorta, 6-, 9-, and 12-week old embryos, adenocarcinoma, osteosarcoma, embryonic carcinoma, and normal umbilical vein cells. The described sequences were compiled from sequences available in GENBANK, and cDNAs generated from human adult and fetal brain, pituitary, hypothalamus, testis, and fetus mRNAs (Edge Biosystems, Gaithersburg, Md.) that were identified using primers generated from human genomic DNA. [0010]
  • The present invention encompasses the nucleotides presented in the Sequence Listing, host cells expressing such nucleotides, the expression products of such nucleotides, and: (a) nucleotides that encode mammalian homologs of the described polynucleotides, including the specifically described NHPs, and the NHP products; (b) nucleotides that encode one or more portions of a NHP that correspond to any functional domain(s), and the polypeptide products specified by such nucleotide sequences, including, but not limited to, the novel regions of any active domain(s); (c) isolated nucleotides that encode mutant versions, engineered or naturally occurring, of the described NHPs, in which all or a part of at least one domain is deleted or altered, and the polypeptide products specified by such nucleotide sequences, including, but not limited to, soluble proteins and peptides; (d) nucleotides that encode chimeric fusion proteins containing all or a portion of a coding region of a NHP, or one of its domains (e.g., a receptor/ligand binding domain, accessory protein/self-association domain, etc.) fused to another peptide or polypeptide; or (e) therapeutic or diagnostic derivatives of the described polynucleotides, such as oligonucleotides, antisense polynucleotides, ribozymes, dsRNA, or gene therapy constructs, comprising a sequence first disclosed in the Sequence Listing. [0011]
  • As discussed above, the present invention includes the human DNA sequences presented in the Sequence Listing (and vectors comprising the same), and additionally contemplates any nucleotide sequence encoding a contiguous NHP open reading frame (ORF) that hybridizes to a complement of a DNA sequence presented in the Sequence Listing under highly stringent conditions, e.g., hybridization to filter-bound DNA in 0.5 M NaHPO[0012] 4, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65° C., and washing in 0.1x SSC/0.1% SDS at 68° C. (Ausubel et al., eds., 1989, Current Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc., and John Wiley & Sons, Inc., N.Y., at p. 2.10.3) and encodes a functionally equivalent expression product. Additionally contemplated are any nucleotide sequences that hybridize to the complement of a DNA sequence that encodes and expresses an amino acid sequence presented in the Sequence Listing under moderately stringent conditions, e.g., washing in 0.2x SSC/0.1% SDS at 42° C. (Ausubel et al., 1989, supra), yet still encode a functionally equivalent NHP product. Functional equivalents of a NHP include naturally occurring NHPs present in other species, and mutant NHPs, whether naturally occurring or engineered (by site directed mutagenesis, gene shuffling, directed evolution as described in, for example, U.S. Pat. Nos. 5,837,458 or 5,723,323, both of which are herein incorporated by reference). The invention also includes degenerate nucleic acid variants of the disclosed NHP polynucleotide sequences.
  • Additionally contemplated are polynucleotides encoding NHP ORFs, or their functional equivalents, encoded by polynucleotide sequences that are about 99, 95, 90, or about 85 percent similar to corresponding regions of SEQ ID NOS:1, 3 or 5 (as measured by BLAST sequence comparison analysis using, for example, the GCG sequence analysis package, as described herein, using default parameters). [0013]
  • The invention also includes nucleic acid molecules, preferably DNA molecules, that hybridize to, and are therefore the complements of, the described NHP-encoding polynucleotides. Such hybridization conditions can be highly stringent or less highly stringent, as described herein. In instances where the nucleic acid molecules are deoxyoligonucleotides (“DNA oligos”), such molecules are generally about 16 to about 100 bases long, or about 20 to about 80 bases long, or about 34 to about 45 bases long, or any variation or combination of sizes represented therein that incorporate a contiguous region of sequence first disclosed in the Sequence Listing. Such oligonucleotides can be used in conjunction with the polymerase chain reaction (PCR) to screen libraries, isolate clones, and prepare cloning and sequencing templates, etc. [0014]
  • Alternatively, such NHP oligonucleotides can be used as hybridization probes for screening libraries, and assessing gene expression patterns (particularly using a microarray or high-throughput “chip” format). Additionally, a series of NHP oligonucleotide sequences, or the complements thereof, can be used to represent all or a portion of the described NHP sequences. An oligonucleotide or polynucleotide sequence first disclosed in at least a portion of one or more of the sequences of SEQ ID NOS:1-6 can be used as a hybridization probe, in conjunction with a solid support matrix/substrate (resins, beads, membranes, plastics, polymers, metal or metallized substrates, crystalline or polycrystalline substrates, etc.). Of particular note are spatially addressable arrays (i.e., gene chips, microtiter plates, etc.) of oligonucleotides and polynucleotides, or corresponding oligopeptides and polypeptides, wherein at least one of the biopolymers present on the spatially addressable array comprises an oligonucleotide or polynucleotide sequence first disclosed in at least one of the sequences of SEQ ID NOS:1-6, or an amino acid sequence encoded thereby. Methods for attaching biopolymers to, or synthesizing biopolymers on, solid support matrices, and conducting binding studies thereon, are disclosed in, inter alia, U.S. Pat. Nos. 5,700,637, 5,556,752, 5,744,305, 4,631,211, 5,445,934, 5,252,743, 4,713,326, 5,424,186, and 4,689,405, the disclosures of which are herein incorporated by reference in their entirety. [0015]
  • Addressable arrays comprising sequences first disclosed in SEQ ID NOS:1-6 can be used to identify and characterize the temporal and tissue specific expression of a gene. These addressable arrays incorporate oligonucleotide sequences of sufficient length to confer the required specificity, yet be within the limitations of the production technology. The length of these probes is usually within a range of between about 8 to about 2000 nucleotides. Preferably the probes consist of 60 nucleotides, and more preferably 25 nucleotides, from the sequences first disclosed in SEQ ID NOS:1-6. [0016]
  • For example, a series of NHP oligonucleotide sequences, or the complements thereof, can be used in chip format to represent all or a portion of the described sequences. The oligonucleotides, typically between about 16 to about 40 (or any whole number within the stated range) nucleotides in length, can partially overlap each other, and/or the sequence may be represented using oligonucleotides that do not overlap. Accordingly, the described polynucleotide sequences shall typically comprise at least about two or three distinct oligonucleotide sequences of at least about 8 nucleotides in length that are each first disclosed in the described Sequence Listing. Such oligonucleotide sequences can begin at any nucleotide present within a sequence in the Sequence Listing, and proceed in either a sense (5′-to-3′) orientation vis-a-vis the described sequence or in an antisense orientation. [0017]
  • Microarray-based analysis allows the discovery of broad patterns of genetic activity, providing new understanding of gene functions, and generating novel and unexpected insight into transcriptional processes and biological mechanisms. The use of addressable arrays comprising sequences first disclosed in SEQ ID NOS:1-6 provides detailed information about transcriptional changes involved in a specific pathway, potentially leading to the identification of novel components, or gene functions that manifest themselves as novel phenotypes. [0018]
  • Probes consisting of sequences first disclosed in SEQ ID NOS:1-6 can also be used in the identification, selection, and validation of novel molecular targets for drug discovery. The use of these unique sequences permits the direct confirmation of drug targets, and recognition of drug dependent changes in gene expression that are modulated through pathways distinct from the intended target of the drug. These unique sequences therefore also have utility in defining and monitoring both drug action and toxicity. [0019]
  • As an example of utility, the sequences first disclosed in SEQ ID NOS:1-6 can be utilized in microarrays, or other assay formats, to screen collections of genetic material from patients who have a particular medical condition. These investigations can also be carried out using the sequences first disclosed in SEQ ID NOS:1-6 in silico, and by comparing previously collected genetic databases and the disclosed sequences using computer software known to those in the art. [0020]
  • Thus the sequences first disclosed in SEQ ID NOS:1-6 can be used to identify mutations associated with a particular disease, and also in diagnostic or prognostic assays. [0021]
  • Although the presently described sequences have been specifically described using nucleotide sequence, it should be appreciated that each of the sequences can uniquely be described using any of a wide variety of additional structural attributes, or combinations thereof. For example, a given sequence can be described by the net composition of the nucleotides present within a given region of the sequence, in conjunction with the presence of one or more specific oligonucleotide sequence(s) first disclosed in SEQ ID NOS:1-6. Alternatively, a restriction map specifying the relative positions of restriction endonuclease digestion sites, or various palindromic or other specific oligonucleotide sequences, can be used to structurally describe a given sequence. Such restriction maps, which are typically generated by widely available computer programs (e.g., the University of Wisconsin GCG sequence analysis package, SEQUENCHER 3.0, Gene Codes Corp., Ann Arbor, Mich., etc.), can optionally be used in conjunction with one or more discrete nucleotide sequence(s) present in the sequence that can be described by the relative position of the sequence relative to one or more additional sequence(s) or one or more restriction sites present in the disclosed sequence. [0022]
  • For oligonucleotide probes, highly stringent conditions may refer, e.g., to washing in 6x SSC/0.05% sodium pyrophosphate at 37° C. (for 14-base oligos), 48° C. (for 17-base oligos), 55° C. (for 20-base oligos), and 60° C. (for 23-base oligos). These nucleic acid molecules may encode or act as NHP antisense molecules, useful, for example, in NHP gene regulation and/or as antisense primers in amplification reactions of NHP nucleic acid sequences. With respect to NHP gene regulation, such techniques can be used to regulate biological functions. Further, such sequences can be used as part of ribozyme and/or triple helix sequences that are also useful for NHP gene regulation. [0023]
  • Inhibitory antisense or double stranded oligonucleotides can additionally comprise at least one modified base moiety that is selected from the group including, but not limited to, 5-fluorouracil, 5,-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. [0024]
  • The antisense oligonucleotide can also comprise at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2-fluoroarabinose, xylulose, and hexose. [0025]
  • In yet another embodiment, the antisense oligonucleotide will comprise at least one modified phosphate backbone selected from the group including, but not limited to, a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or analog thereof. [0026]
  • In yet another embodiment, the antisense oligonucleotide is an α-anomeric oligonucleotide. An α-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gautier et al., 1987, Nucl. Acids Res. 15:6625-6641). The oligonucleotide is a 2′-0-methylribonucleotide (Inoue et al., 1987, Nucl. Acids Res. 15:6131-6148), or a chimeric RNA-DNA analogue (Inoue et al., 1987, FEBS Lett. 215:327-330). Alternatively, double stranded RNA can be used to disrupt the expression and function of a targeted NHP. [0027]
  • Oligonucleotides of the invention can be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (such as are commercially available from Biosearch, Applied Biosystems, etc.). As examples, phosphorothioate oligonucleotides can be synthesized by the method of Stein et al. (1988, Nucl. Acids Res. 16:3209), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., 1988, Proc. Natl. Acad. Sci. USA 85:7448-7451), etc. [0028]
  • Low stringency conditions are well-known to those of skill in the art, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived. For guidance regarding such conditions see, for example, Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (and periodic updates thereof), and Ausubel et al., 1989, supra. [0029]
  • Alternatively, suitably labeled NHP nucleotide probes can be used to screen a human genomic library using appropriately stringent conditions or by PCR. The identification and characterization of human genomic clones is helpful for identifying polymorphisms (including, but not limited to, nucleotide repeats, microsatellite alleles, single nucleotide polymorphisms, or coding single nucleotide polymorphisms), determining the genomic structure of a given locus/allele, and designing diagnostic tests. For example, sequences derived from regions adjacent to the intron/exon boundaries of the human gene can be used to design primers for use in amplification assays to detect mutations within the exons, introns, splice sites (e.g., splice acceptor and/or donor sites), etc., that can be used in diagnostics and pharmacogenomics. [0030]
  • For example, the present sequences can be used in restriction fragment length polymorphism (RFLP) analysis to identify specific individuals. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification (as generally described in U.S. Pat. No. 5,272,057, incorporated herein by reference). In addition, the sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another “identification marker” (i.e., another DNA sequence that is unique to a particular individual). Actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments. [0031]
  • Further, a NHP homolog can be isolated from nucleic acid from an organism of interest by performing PCR using two degenerate or “wobble” oligonucleotide primer pools designed on the basis of amino acid sequences within the NHP products disclosed herein. The template for the reaction may be total RNA, mRNA, genomic DNA and/or cDNA obtained by reverse transcription of mRNA prepared from, for example, human or non-human cell lines or tissue known to express, or suspected of expressing, an allele of a NHP gene. [0032]
  • The PCR product can be subcloned and sequenced to ensure that the amplified sequences represent the sequence of the desired NHP gene. The PCR fragment can then be used to isolate a full length cDNA clone by a variety of methods. For example, the amplified fragment can be labeled and used to screen a cDNA library, such as a bacteriophage cDNA library. Alternatively, the labeled fragment can be used to isolate genomic clones via the screening of a genomic library. [0033]
  • PCR technology can also be used to isolate full length cDNA sequences. For example, RNA can be isolated, following standard procedures, from an appropriate cellular or tissue source (i.e., one known to express, or suspected of expressing, a NHP gene). A reverse transcription (RT) reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5′ end of the amplified fragment for the priming of first strand synthesis. The resulting RNA/DNA hybrid may then be “tailed” using a standard terminal transferase reaction, the hybrid may be digested with RNase H, and second strand synthesis may then be primed with a complementary primer. Thus, cDNA sequences upstream of the amplified fragment can be isolated. For a review of cloning strategies that can be used, see, e.g., Sambrook et al., 1989, supra. [0034]
  • A cDNA encoding a mutant NHP sequence can be isolated, for example, by using PCR. In this case, the first cDNA strand may be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue known to express, or suspected of expressing, a NHP, in an individual putatively carrying a mutant NHP allele, and by extending the new strand with reverse transcriptase. The second strand of the cDNA is then synthesized using an oligonucleotide that hybridizes specifically to the 5′ end of the normal sequence. Using these two primers, the product is then amplified via PCR, optionally cloned into a suitable vector, and subjected to DNA sequence analysis through methods well-known to those of skill in the art. By comparing the DNA sequence of the mutant NHP allele to that of a corresponding normal NHP allele, the mutation(s) responsible for the loss or alteration of function of the mutant NHP gene product can be ascertained. [0035]
  • Alternatively, a genomic library can be constructed using DNA obtained from an individual suspected of carrying, or known to carry, a mutant NHP allele (e.g., a person manifesting a NHP-associated phenotype such as, for example, behavioral disorders, immune disorders, obesity, high blood pressure, etc.), or a cDNA library can be constructed using RNA from a tissue known to express, or suspected of expressing, a mutant NHP allele. A normal NHP gene, or any suitable fragment thereof, can then be labeled and used as a probe to identify the corresponding mutant NHP allele in such libraries. Clones containing mutant NHP sequences can then be purified and subjected to sequence analysis according to methods well-known to those skilled in the art. [0036]
  • Additionally, an expression library can be constructed utilizing cDNA synthesized from, for example, RNA isolated from a tissue known to express, or suspected of expressing, a mutant NHP allele in an individual suspected of carrying, or known to carry, such a mutant allele. In this manner, gene products made by the putatively mutant tissue may be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against a normal NHP product, as described below (for screening techniques, see, for example, Harlow and Lane, eds., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Press, Cold Spring Harbor, N.Y.). [0037]
  • Additionally, screening can be accomplished by screening with labeled NHP fusion proteins, such as, for example, alkaline phosphatase-NHP or NHP-alkaline phosphatase fusion proteins. In cases where a NHP mutation results in an expression product with altered function (e.g., as a result of a missense or a frameshift mutation), polyclonal antibodies to a NHP are likely to cross-react with a corresponding mutant NHP expression product. Library clones detected via their reaction with such labeled antibodies can be purified and subjected to sequence analysis according to methods well-known in the art. [0038]
  • An additional application of the described novel human polynucleotide sequences is their use in the molecular mutagenesis/evolution of proteins that are at least partially encoded by the described novel sequences using, for example, polynucleotide shuffling or related methodologies. Such approaches are described in U.S. Pat. Nos. 5,830,721, 5,837,458, 6,117,679, and 5,723,323, which are herein incorporated by reference in their entirety. [0039]
  • The invention also encompasses: (a) DNA vectors that contain any of the foregoing NHP coding sequences and/or their complements (i.e., antisense); (b) DNA expression vectors that contain any of the foregoing NHP coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences (for example, baculovirus as described in U.S. Pat. No. 5,869,336 herein incorporated by reference); (c) genetically engineered host cells that contain any of the foregoing NHP coding sequences operatively associated with a regulatory element that directs the expression of the coding sequences in the host cell; and (d) genetically engineered host cells that express an endogenous NHP sequence under the control of an exogenously introduced regulatory element (i.e., gene activation). As used herein, regulatory elements include, but are not limited to, inducible and non-inducible promoters, enhancers, operators, and other elements known to those skilled in the art that drive and regulate expression. Such regulatory elements include, but are not limited to, the cytomegalovirus (hCMV) immediate early gene, regulatable, viral elements (particularly retroviral LTR promoters), the early or late promoters of SV40 or adenovirus, the lac system, the trp system, the TAC system, the TRC system, the major operator and promoter regions of phage lambda, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase (PGK), the promoters of acid phosphatase, and the promoters of the yeast α-mating factors. [0040]
  • Where, as in the present instance, some of the described NHP peptides or polypeptides are thought to be cytoplasmic or nuclear proteins (although processed forms or fragments can be secreted or membrane associated), expression systems can be engineered that produce soluble derivatives of a NHP (corresponding to a NHP extracellular and/or intracellular domains, or truncated polypeptides lacking one or more hydrophobic domains) and/or NHP fusion protein products (especially NHP-Ig fusion proteins, i.e., fusions of a NHP domain to an IgFc), NHP antibodies, and anti-idiotypic antibodies (including Fab fragments) that can be used in therapeutic applications. Preferably, the above expression systems are engineered to allow the desired peptide or polypeptide to be recovered from the culture media. [0041]
  • The present invention also encompasses antibodies and anti-idiotypic antibodies (including Fab fragments), antagonists and agonists of a NHP, as well as compounds or nucleotide constructs that inhibit expression of a NHP sequence (transcription factor inhibitors, antisense and ribozyme molecules, or open reading frame sequence or regulatory sequence replacement constructs), or promote the expression of a NHP (e.g., expression constructs in which NHP coding sequences are operatively associated with expression control elements such as promoters, promoter/enhancers, etc.). [0042]
  • The NHPs or NHP peptides, NHP fusion proteins, NHP nucleotide sequences, antibodies, antagonists and agonists can be useful for the detection of mutant NHPs, or inappropriately expressed NHPs, for the diagnosis of disease. The NHP proteins or peptides, NHP fusion proteins, NHP nucleotide sequences, host cell expression systems, antibodies, antagonists, agonists and genetically engineered cells and animals can be used for screening for drugs (or high throughput screening of combinatorial libraries) effective in the treatment of the symptomatic or phenotypic manifestations of perturbing the normal function of a NHP in the body. The use of engineered host cells and/or animals can offer an advantage in that such systems allow not only for the identification of compounds that bind to the endogenous receptor/ligand of a NHP, but can also identify compounds that trigger NHP-mediated activities or pathways. [0043]
  • Finally, the NHP products can be used as therapeutics. For example, soluble derivatives such as NHP peptides/domains corresponding to NHPs, NHP fusion protein products (especially NHP-Ig fusion proteins, i.e., fusions of a NHP, or a domain of a NHP, to an IgFc), NHP antibodies and anti-idiotypic antibodies (including Fab fragments), antagonists or agonists (including compounds that modulate or act on downstream targets in a NHP-mediated pathway) can be used to directly treat diseases or disorders. For instance, the administration of an effective amount of a soluble NHP, a NHP-IgFc fusion protein, or an anti-idiotypic antibody (or its Fab) that mimics the NHP, could activate or effectively antagonize the endogenous NHP or a protein interactive therewith. Nucleotide constructs encoding such NHP products can be used to genetically engineer host cells to express such products in vivo; these genetically engineered cells function as “bioreactors” in the body delivering a continuous supply of a NHP, a NHP peptide, or a NHP fusion protein to the body. Nucleotide constructs encoding functional NHPs, mutant NHPs, as well as antisense and ribozyme molecules, can also be used in “gene therapy” approaches for the modulation of NHP expression. Thus, the invention also encompasses pharmaceutical formulations and methods for treating biological disorders. [0044]
  • Various aspects of the invention are described in greater detail in the subsections below. [0045]
  • THE NHP SEQUENCES
  • The cDNA sequences (SEQ ID NOS:1, 3, and 5) and corresponding deduced amino acid sequences (SEQ ID NOS:2, 4, and 6, respectively) of the described NHPs are presented in the Sequence Listing. [0046]
  • Expression analysis has provided evidence that the described NHPs can be expressed in a moderate range of human tissues. In addition to serine-threonine kinases, the described NHPs also share significant similarity to a range of additional kinase families, including kinases associated with signal transduction, from a variety of phyla and species. The genomic locus encoding the described NHPs is apparently encoded on human chromosome 19 (see GENBANK accession no. AC020922). As such, the described sequences are useful, inter alia, for mapping the coding regions of the human genome, and particularly chromosome 19. [0047]
  • Given the physiological importance of protein kinases, they have been subject to intense scrutiny, as exemplified and discussed in U.S. Pat. Nos. 5,756,289 and 5,817,479, herein incorporated by reference in their entirety, which describe uses and utilities that are applicable to the described NHPs. [0048]
  • NHP gene products can also be expressed in transgenic animals. Animals of any species, including, but not limited to, worms, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, birds, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, may be used to generate NHP transgenic animals. [0049]
  • Any technique known in the art may be used to introduce a NHP transgene into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (Hoppe and Wagner, 1989, U.S. Pat. No. 4,873,191); retrovirus-mediated gene transfer into germ lines (Van der Putten et al., 1985, Proc. Natl. Acad. Sci. USA 82:6148-6152); gene targeting in embryonic stem cells (Thompson et al., 1989, Cell 56:313-321); electroporation of embryos (Lo, 1983, Mol Cell. Biol. 3:1803-1814); and sperm-mediated gene transfer (Lavitrano et al., 1989, Cell 57:717-723); etc. For a review of such techniques, see Gordon, 1989, Transgenic Animals, Intl. Rev. Cytol. 115:171-229, which is incorporated by reference herein in its entirety. [0050]
  • The present invention provides for transgenic animals that carry a NHP transgene in all their cells, as well as animals that carry a transgene in some, but not all their cells, i.e., mosaic animals or somatic cell transgenic animals. A transgene may be integrated as a single transgene, or in concatamers, e.g., head-to-head tandems or head-to-tail tandems. A transgene may also be selectively introduced into and activated in a particular cell-type by following, for example, the teaching of Lasko et al., 1992, Proc. Natl. Acad. Sci. USA 89:6232-6236. The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell-type of interest, and will be apparent to those of skill in the art. [0051]
  • When it is desired that a NHP transgene be integrated into the chromosomal site of the endogenous NHP gene, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous NHP gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous NHP gene (i.e., “knockout” animals). [0052]
  • The transgene can also be selectively introduced into a particular cell-type, thus inactivating the endogenous NHP gene in only that cell-type, by following, for example, the teaching of Gu et al., 1994, Science 265:103-106. The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell-type of interest, and will be apparent to those of skill in the art. [0053]
  • Once transgenic animals have been generated, the expression of the recombinant NHP sequence may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals may also be assessed using techniques that include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and RT-PCR. Samples of NHP gene-expressing tissue may also be evaluated immunocytochemically using antibodies specific for the NHP transgene product. [0054]
  • The present invention also provides for “knock-in” animals. Knock-in animals are those in which a polynucleotide sequence (i.e., a gene or a cDNA) that the animal does not naturally have in its genome is inserted in such a way that it is expressed. Examples include, but are not limited to, a human gene or cDNA used to replace its murine ortholog in the mouse, a murine cDNA used to replace the murine gene in the mouse, and a human gene or cDNA or murine cDNA that is tagged with a reporter construct used to replace the murine ortholog or gene in the mouse. Such replacements can occur at the locus of the murine ortholog or gene, or at another specific site. Such knock-in animals are useful for the in vivo study, testing and validation of, intra alia, human drug targets, as well as for compounds that are directed at the same, and therapeutic proteins. [0055]
  • NHPS AND NHP POLYPEPTIDES
  • NHPs, NHP polypeptides, NHP peptide fragments, mutated, truncated, or deleted forms of the NHPs, and/or NHP fusion proteins can be prepared for a variety of uses. These uses include, but are not limited to, the generation of antibodies, as reagents in diagnostic assays, for the identification of other cellular gene products related to a NHP, and as reagents in assays for screening for compounds that can be used as pharmaceutical reagents useful in the therapeutic treatment of mental, biological, or medical disorders and diseases. Given the similarity information and expression data, the described NHPs can be targeted (by drugs, oligonucleotides, antibodies, etc.) in order to treat disease, or to therapeutically augment the efficacy of therapeutic agents. [0056]
  • The Sequence Listing discloses the amino acid sequences encoded by the described NHP-encoding polynucleotides. The NHPs display initiator methionines that are present in DNA sequence contexts consistent with eucaryotic translation initiation sites. The NHPs do not display consensus signal sequences, which indicates that they may be cytoplasmic or possibly nuclear proteins, although they may also be secreted or membrane associated (given the presence of several hydrophobic domains). [0057]
  • The NHP amino acid sequences of the invention include the amino acid sequences presented in the Sequence Listing, as well as analogues and derivatives thereof. Further, corresponding NHP homologues from other species are encompassed by the invention. In fact, any NHP protein encoded by the NHP nucleotide sequences described herein are within the scope of the invention, as are any novel polynucleotide sequences encoding all or any novel portion of an amino acid sequence presented in the Sequence Listing. The degenerate nature of the genetic code is well-known, and, accordingly, each amino acid presented in the Sequence Listing is generically representative of the well-known nucleic acid “triplet” codon, or in many cases codons, that can encode the amino acid. As such, as contemplated herein, the amino acid sequences presented in the Sequence Listing, when taken together with the genetic code (see, for example, Table 4-1 at page 109 of “Molecular Cell Biology”, 1986, J. Darnell et al., eds., Scientific American Books, New York, N.Y., herein incorporated by reference), are generically representative of all the various permutations and combinations of nucleic acid sequences that can encode such amino acid sequences. [0058]
  • The invention also encompasses proteins that are functionally equivalent to the NHPs encoded by the presently described nucleotide sequences, as judged by any of a number of criteria, including, but not limited to, the ability to bind and modify a NHP substrate, or the ability to effect an identical or complementary downstream pathway, or a change in cellular metabolism (e.g., proteolytic activity, ion flux, phosphorylation, etc.). Such functionally equivalent NHP proteins include, but are not limited to, additions or substitutions of amino acid residues within the amino acid sequence encoded by the NHP nucleotide sequences described herein, but that result in a silent change, thus producing a functionally equivalent expression product. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. [0059]
  • A variety of host-expression vector systems can be used to express the NHP nucleotide sequences of the invention. Where the NHP peptide or polypeptide can exist, or has been engineered to exist, as a soluble or secreted molecule, the soluble NHP peptide or polypeptide can be recovered from the culture media. Such expression systems also encompass engineered host cells that express a NHP, or functional equivalent, in situ. Purification or enrichment of a NHP from such expression systems can be accomplished using appropriate detergents and lipid micelles and methods well-known to those skilled in the art. However, such engineered host cells themselves may be used in situations where it is important not only to retain the structural and functional characteristics of a NHP, but to assess biological activity, e.g., in certain drug screening assays. [0060]
  • The expression systems that may be used for purposes of the invention include, but are not limited to, microorganisms such as bacteria (e.g., [0061] E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing NHP nucleotide sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing NHP nucleotide sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing NHP nucleotide sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing NHP nucleotide sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3) harboring recombinant expression constructs containing NHP nucleotide sequences and promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
  • In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the NHP product being expressed. For example, when a large quantity of such a protein is to be produced for the generation of pharmaceutical compositions of or containing a NHP, or for raising antibodies to a NHP, vectors that direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited to, the [0062] E. coli expression vector pUR278 (Ruther et al., 1983, EMBO J. 2:1791), in which a NHP coding sequence may be ligated individually into the vector in-frame with the lacZ coding region so that a fusion protein is produced; pIN vectors (Inouye and Inouye, 1985, Nucleic Acids Res. 13:3101-3109; Van Heeke and Schuster, 1989, J. Biol. Chem. 264:5503-5509); and the like. PGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target expression product can be released from the GST moiety.
  • In an exemplary insect system, [0063] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign polynucleotide sequences. The virus grows in Spodoptera frugiperda cells. A NHP coding sequence can be cloned individually into a non-essential region (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter). Successful insertion of a NHP coding sequence will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e., virus lacking the proteinaceous coat coded for by the polyhedrin gene). These recombinant viruses are then used to infect Spodoptera frugiperda cells in which the inserted sequence is expressed (e.g., see Smith et al., 1983, J. Virol. 46:584; Smith, U.S. Pat. No. 4,215,051).
  • In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the NHP nucleotide sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric sequence may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E[0064] 1 or E3) will result in a recombinant virus that is viable and capable of expressing a NHP product in infected hosts (e.g., see Logan and Shenk, 1984, Proc. Natl. Acad. Sci. USA 81:3655-3659). Specific initiation signals may also be required for efficient translation of inserted NHP nucleotide sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire NHP gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of a NHP coding sequence is inserted, exogenous translational control signals, including, perhaps, the ATG initiation codon, may be provided. Furthermore, the initiation codon should be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bitter et al., 1987, Methods in Enzymol. 153:516-544).
  • In addition, a host cell strain may be chosen that modulates the expression of the inserted sequences, or modifies and processes the expression product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and expression products. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. To this end, eukaryotic host cells that possess the cellular machinery for the desired processing of the primary transcript, glycosylation, and phosphorylation of the expression product may be used. Such mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, WI38, and in particular, human cell lines. [0065]
  • For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines that stably express the NHP sequences described herein can be engineered. Rather than using expression vectors that contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci, which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines that express the NHP product. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the NHP product. [0066]
  • A number of selection systems may be used, including, but not limited to, the herpes simplex virus thymidine kinase (Wigler et al., 1977, Cell 11:223), hypoxanthine-guanine phosphoribosyltransferase (Szybalska and Szybalski, 1962, Proc. Natl. Acad. Sci. USA 48:2026), and adenine, phosphoribosyltransferase (Lowy et al., 1980, Cell 22:817) genes, which can be employed in tk[0067] , hgprt or aprt cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., 1980, Proc. Natl. Acad. Sci. USA 77:3567; O'Hare et al., 1981, Proc. Natl. Acad. Sci. USA 78:1527); gpt, which confers resistance to mycophenolic acid (Mulligan and Berg, 1981, Proc. Natl. Acad. Sci. USA 78:2072); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., 1981, J. Mol. Biol. 150:1); and hygro, which confers resistance to hygromycin (Santerre et al., 1984, Gene 30:147).
  • Alternatively, any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed. Another exemplary system allows for the ready purification of non-denatured fusion proteins expressed in human cell lines (Janknecht et al., 1991, Proc. Natl. Acad. Sci. USA 88:8972-8976). In this system, the sequence of interest is subcloned into a vaccinia recombination plasmid such that the sequence's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni[0068] 2+·nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • Also encompassed by the present invention are fusion proteins that direct a NHP to a target organ and/or facilitate transport across the membrane into the cytosol. Conjugation of NHPs to antibody molecules or their Fab fragments could be used to target cells bearing a particular epitope. Attaching an appropriate signal sequence to a NHP would also transport a NHP to a desired location within the cell. Alternatively targeting of a NHP or its nucleic acid sequence might be achieved using liposome or lipid complex based delivery systems. Such technologies are described in “Liposomes: A Practical Approach”, New, R.R.C., ed., Oxford University Press, N.Y., and in U.S. Pat. Nos. 4,594,595, 5,459,127, 5,948,767 and 6,110,490 and their respective disclosures, which are herein incorporated by reference in their entirety. Additionally embodied are novel protein constructs engineered in such a way that they facilitate transport of NHPs to a target site or desired organ, where they cross the cell membrane and/or the nucleus where the NHPs can exert their functional activity. This goal may be achieved by coupling of a NHP to a cytokine or other ligand that provides targeting specificity, and/or to a protein transducing domain (see generally U.S. Provisional Patent Application Ser. Nos. 60/111,701 and 60/056,713, both of which are herein incorporated by reference, for examples of such transducing sequences), to facilitate passage across cellular membranes, and can optionally be engineered to include nuclear localization signals. [0069]
  • Additionally contemplated are oligopeptides that are modeled on an amino acid sequence first described in the Sequence Listing. Such NHP oligopeptides are generally between about 10 to about 100 amino acids long, or between about 16 to about 80 amino acids long, or between about 20 to about 35 amino acids long, or any variation or combination of sizes represented therein that incorporate a contiguous region of sequence first disclosed in the Sequence Listing. Such NHP oligopeptides can be of any length disclosed within the above ranges and can initiate at any amino acid position represented in the Sequence Listing. [0070]
  • The invention also contemplates “substantially isolated” or “substantially pure” proteins or polypeptides. By a “substantially isolated” or “substantially pure” protein or polypeptide is meant a protein or polypeptide that has been separated from at least some of those components which naturally accompany it. Typically, the protein or polypeptide is substantially isolated or pure when it is at least 60%, by weight, free from the proteins and other naturally-occurring organic molecules with which it is naturally associated in vivo. Preferably, the purity of the preparation is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight. A substantially isolated or pure protein or polypeptide may be obtained, for example, by extraction from a natural source, by expression of a recombinant nucleic acid encoding the protein or polypeptide, or by chemically synthesizing the protein or polypeptide. [0071]
  • Purity can be measured by any appropriate method, e.g., column chromatography such as immunoaffinity chromatography using an antibody specific for the protein or polypeptide, polyacrylamide gel electrophoresis, or HPLC analysis. A protein or polypeptide is substantially free of naturally associated components when it is separated from at least some of those contaminants which accompany it in its natural state. Thus, a polypeptide which is chemically synthesized or produced in a cellular system different from the cell from which it naturally originates will be, by definition, substantially free from its naturally associated components. Accordingly, substantially isolated or pure proteins or polypeptides include eukaryotic proteins synthesized in [0072] E. coli, other prokaryotes, or any other organism in which they do not naturally occur.
  • ANTIBODIES TO NHP PRODUCTS
  • Antibodies that specifically recognize one or more epitopes of a NHP, epitopes of conserved variants of a NHP, or peptide fragments of a NHP, are also encompassed by the invention. Such antibodies include, but are not limited to, polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′)[0073] 2 fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • The antibodies of the invention can be used, for example, in the detection of a NHP in a biological sample and may, therefore, be utilized as part of a diagnostic or prognostic technique whereby patients may be tested for abnormal amounts of a NHP. Such antibodies may also be utilized in conjunction with, for example, compound screening schemes for the evaluation of the effect of test compounds on expression and/or activity of a NHP expression product. Additionally, such antibodies can be used in conjunction with gene therapy to, for example, evaluate normal and/or engineered NHP-expressing cells prior to their introduction into a patient. Such antibodies may additionally be used in methods for the inhibition of abnormal NHP activity. Thus, such antibodies may be utilized as a part of treatment methods. [0074]
  • For the production of antibodies, various host animals may be immunized by injection with a NHP, a NHP peptide (e.g., one corresponding to a functional domain of a NHP), truncated NHP polypeptides (NHP in which one or more domains have been deleted), functional equivalents of a NHP, or mutated variants of a NHP. Such host animals may include, but are not limited to, pigs, rabbits, mice, goats, and rats, to name but a few. Various adjuvants may be used to increase the immunological response, depending on the host species, including, but not limited to, Freund's adjuvant (complete and incomplete), mineral salts such as aluminum hydroxide or aluminum phosphate, chitosan, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and [0075] Corynebacterium parvum. Alternatively, the immune response could be enhanced by combination and/or coupling with molecules such as keyhole limpet hemocyanin, tetanus toxoid, diphtheria toxoid, ovalbumin, cholera toxin, or fragments thereof. Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of the immunized animals.
  • Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler and Milstein, (1975, Nature 256:495-497; and U.S. Pat. No. 4,376,110), the human B-cell hybridoma technique (Kosbor et al., 1983, Immunology Today 4:72; Cole et al., 1983, Proc. Natl. Acad. Sci. USA 80:2026-2030), and the EBV-hybridoma technique (Cole. et al., 1985, Monoclonal Antibodies And Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Such antibodies may be of any immunoglobulin class, including IgG, IgM, IgE, IgA, and IgD, and any subclass thereof. The hybridomas producing the mAbs of this invention may be cultivated in vitro or in vivo. Production of high titers of mAbs in vivo makes this the presently preferred method of production. [0076]
  • In addition, techniques developed for the production of “chimeric antibodies” (Morrison et al., 1984, Proc. Natl. Acad. Sci. USA 81:6851-6855; Neuberger. et al., 1984, Nature, 312:604-608; Takeda et al., 1985, Nature, 314:452-454), by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity, can be used. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region. Such technologies are described in U.S. Pat. Nos. 6,114,598, 6,075,181 and 5,877,397 and their respective disclosures, which are herein incorporated by reference in their entirety. Also encompassed by the present invention is the use of fully humanized monoclonal antibodies, as described in U.S. Pat. No. 6,150,584 and respective disclosures, which are herein incorporated by reference in their entirety. [0077]
  • Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; Bird, 1988, Science 242:423-426; Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; and Ward et al., 1989, Nature 341:544-546) can be adapted to produce single chain antibodies against NHP expression products. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. [0078]
  • Antibody fragments that recognize specific epitopes may be generated by known techniques. For example, such fragments include, but are not limited to: F(ab′)[0079] 2 fragments, which can be produced by pepsin digestion of an antibody molecule; and Fab fragments, which can be generated by reducing the disulfide bridges of F(ab′)2 fragments. Alternatively, Fab expression libraries may be constructed (Huse et al., 1989, Science, 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity.
  • Antibodies to a NHP can, in turn, be utilized to generate anti-idiotype antibodies that “mimic” a given NHP, using techniques well-known to those skilled in the art (see, e.g., Greenspan and Bona, 1993, FASEB J. 7:437-444; and Nissinoff, 1991, J. Immunol. 147:2429-2438). For example, antibodies that bind to a NHP domain and competitively inhibit the binding of a NHP to its cognate receptor/ligand can be used to generate anti-idiotypes that “mimic” the NHP and, therefore, bind, activate, or neutralize a NHP, NHP receptor, or NHP ligand. Such anti-idiotypic antibodies or Fab fragments of such anti-idiotypes can be used in therapeutic regimens involving a NHP-mediated pathway. [0080]
  • Additionally given the high degree of relatedness of mammalian NHPS, the presently described knock-out mice (having never seen a NHP, and thus never been tolerized to a NHP) have a unique utility, as they can be advantageously applied to the generation of antibodies against the disclosed mammalian NHPs (i.e., a NHP will be immunogenic in NHP knock-out animals). [0081]
  • The present invention is not to be limited in scope by the specific embodiments described herein, which are intended as single illustrations of individual aspects of the invention, and functionally equivalent methods and components are within the scope of the invention. Indeed, various modifications of the invention, in addition to those shown and described herein, will become apparent to those skilled in the art from the foregoing description. Such modifications are intended to fall within the scope of the appended claims. All cited publications, patents, and patent applications are herein incorporated by reference in their entirety. [0082]
  • 1 6 1 2337 DNA homo sapiens 1 atgtcgtccg gggccaagga gggaggtggg ggctctcccg cctaccacct cccccacccc 60 cacccccacc caccccagca cgcccaatat gtgggcccct atcggctgga gaagacgctg 120 ggcaaaggac agacagggct ggttaaactc ggggtccact gcatcacggg tcagaaggtc 180 gccatcaaga tcgtgaaccg ggagaagctg tcggagtcgg tgctgatgaa ggtggagcgg 240 gagatcgcca tcctgaagct catcgaacac ccacatgtcc tcaagctcca cgacgtctac 300 gagaacaaga aatatttgta cctggttctg gagcacgtct cggggggtga gctattcgac 360 tacctggtaa agaaggggag actgacgccc aaggaggccc gaaagttctt ccgccagatt 420 gtgtctgcgc tggacttctg ccacagctac tccatctgcc acagagacct aaagcccgag 480 aacctgcttt tggatgagaa aaacaacatc cgcattgcag acttcggcat ggcgtccctg 540 caggtggggg acagcctcct ggagaccagc tgcgggtccc cccattatgc gtgtccagag 600 gtgattaagg gggaaaaata tgatggccgc cgggcagaca tgtggagctg tggagtcatc 660 ctcttcgccc tgctcgtggg ggctctgccc tttgatgacg acaacctccg ccagctgctg 720 gagaaggtga aacggggcgt cttccacatg ccccacttca ttcctccaga ttgccagagc 780 ctcctgaggg gaatgatcga agtggagccc gaaaaaaggc tcagtctgga gcaaattcag 840 aaacatcctt ggtacctagg cgggaaacac gagccagacc cgtgcctgga gccagcccct 900 ggccgccggg tagccatgcg gagcctgcca tccaacggag agctggaccc cgacgtccta 960 gagagcatgg catcactggg ctgcttcagg gaccgcgaga ggctgcatcg cgagctgcgc 1020 agtgaggagg agaaccaaga aaagatgata tattatctgc ttttggatcg gaaggagcgg 1080 tatcccagct gtgaggacca ggacctgcct ccccggaatg atgttgaccc cccccggaag 1140 cgtgtggatt ctcccatgct gagccgtcac gggaagcggc gaccagagcg gaagtccatg 1200 gaagtcctga gcatcaccga tgccgggggt ggtggctccc ctgtacccac ccgacgggcc 1260 ttggagatgg cccagcacag ccagagatcc cgtagcgtca gtggagcctc cacgggtctg 1320 tcctccagcc ctctaagcag cccaaggagt ccggtctttt ccttttcacc ggagccgggg 1380 gctggagatg aggctcgagg cgggggctcc ccgacttcca aaacgcagac gctgccttct 1440 cggggcccca ggggtggggg cgccggggag cagcccccgc cccccagtgc ccgctccaca 1500 cccctgcccg gccccccagg ctccccgcgc tcctctggcg ggaccccctt gcactcgcct 1560 ctgcacacgc cccgggccag tcccaccggg accccgggga caacaccacc ccccagcccc 1620 ggcggtggcg tcgggggagc cgcctggagg agtcgtctca actccatccg caacagcttc 1680 ctgggctccc ctcgctttca ccggcgcaag atgcaggtcc ctaccgctga ggagatgtcc 1740 agcttgacgc cagagtcctc cccggagctg gcaaaacgct cctggttcgg gaacttcatc 1800 tccttggaca aagaagaaca aatattcctc gtgctaaagg acaaacctct cagcagcatc 1860 aaagcagaca tcgtccatgc ctttctgtcg atccccagcc tgagtcacag tgtgctgtca 1920 cagaccagct tcagggccga gtacaaggcc agtggcggcc cctccgtctt ccaaaagccc 1980 gtccgcttcc aggtggacat cagctcctct gagggtccag agccctcccc gcgacgggac 2040 ggcagcggag gtggtggcat ctactccgtc accttcactc tcatctcggg tcccagccgt 2100 cggttcaagc gagtggtgga gaccatccag gcacagctcc tgagcactca tgaccagccc 2160 tccgtgcagg ccctggcaga cgagaagaac ggggcccaga cccggcctgc tggtgcccca 2220 ccccgaagcc tgcagccccc acccggccgc ccagacccag agctgagcag ctctccccgc 2280 cgaggccccc ccaaggacaa gaagctcctg gccaccaacg ggacccctct gccctga 2337 2 778 PRT homo sapiens 2 Met Ser Ser Gly Ala Lys Glu Gly Gly Gly Gly Ser Pro Ala Tyr His 1 5 10 15 Leu Pro His Pro His Pro His Pro Pro Gln His Ala Gln Tyr Val Gly 20 25 30 Pro Tyr Arg Leu Glu Lys Thr Leu Gly Lys Gly Gln Thr Gly Leu Val 35 40 45 Lys Leu Gly Val His Cys Ile Thr Gly Gln Lys Val Ala Ile Lys Ile 50 55 60 Val Asn Arg Glu Lys Leu Ser Glu Ser Val Leu Met Lys Val Glu Arg 65 70 75 80 Glu Ile Ala Ile Leu Lys Leu Ile Glu His Pro His Val Leu Lys Leu 85 90 95 His Asp Val Tyr Glu Asn Lys Lys Tyr Leu Tyr Leu Val Leu Glu His 100 105 110 Val Ser Gly Gly Glu Leu Phe Asp Tyr Leu Val Lys Lys Gly Arg Leu 115 120 125 Thr Pro Lys Glu Ala Arg Lys Phe Phe Arg Gln Ile Val Ser Ala Leu 130 135 140 Asp Phe Cys His Ser Tyr Ser Ile Cys His Arg Asp Leu Lys Pro Glu 145 150 155 160 Asn Leu Leu Leu Asp Glu Lys Asn Asn Ile Arg Ile Ala Asp Phe Gly 165 170 175 Met Ala Ser Leu Gln Val Gly Asp Ser Leu Leu Glu Thr Ser Cys Gly 180 185 190 Ser Pro His Tyr Ala Cys Pro Glu Val Ile Lys Gly Glu Lys Tyr Asp 195 200 205 Gly Arg Arg Ala Asp Met Trp Ser Cys Gly Val Ile Leu Phe Ala Leu 210 215 220 Leu Val Gly Ala Leu Pro Phe Asp Asp Asp Asn Leu Arg Gln Leu Leu 225 230 235 240 Glu Lys Val Lys Arg Gly Val Phe His Met Pro His Phe Ile Pro Pro 245 250 255 Asp Cys Gln Ser Leu Leu Arg Gly Met Ile Glu Val Glu Pro Glu Lys 260 265 270 Arg Leu Ser Leu Glu Gln Ile Gln Lys His Pro Trp Tyr Leu Gly Gly 275 280 285 Lys His Glu Pro Asp Pro Cys Leu Glu Pro Ala Pro Gly Arg Arg Val 290 295 300 Ala Met Arg Ser Leu Pro Ser Asn Gly Glu Leu Asp Pro Asp Val Leu 305 310 315 320 Glu Ser Met Ala Ser Leu Gly Cys Phe Arg Asp Arg Glu Arg Leu His 325 330 335 Arg Glu Leu Arg Ser Glu Glu Glu Asn Gln Glu Lys Met Ile Tyr Tyr 340 345 350 Leu Leu Leu Asp Arg Lys Glu Arg Tyr Pro Ser Cys Glu Asp Gln Asp 355 360 365 Leu Pro Pro Arg Asn Asp Val Asp Pro Pro Arg Lys Arg Val Asp Ser 370 375 380 Pro Met Leu Ser Arg His Gly Lys Arg Arg Pro Glu Arg Lys Ser Met 385 390 395 400 Glu Val Leu Ser Ile Thr Asp Ala Gly Gly Gly Gly Ser Pro Val Pro 405 410 415 Thr Arg Arg Ala Leu Glu Met Ala Gln His Ser Gln Arg Ser Arg Ser 420 425 430 Val Ser Gly Ala Ser Thr Gly Leu Ser Ser Ser Pro Leu Ser Ser Pro 435 440 445 Arg Ser Pro Val Phe Ser Phe Ser Pro Glu Pro Gly Ala Gly Asp Glu 450 455 460 Ala Arg Gly Gly Gly Ser Pro Thr Ser Lys Thr Gln Thr Leu Pro Ser 465 470 475 480 Arg Gly Pro Arg Gly Gly Gly Ala Gly Glu Gln Pro Pro Pro Pro Ser 485 490 495 Ala Arg Ser Thr Pro Leu Pro Gly Pro Pro Gly Ser Pro Arg Ser Ser 500 505 510 Gly Gly Thr Pro Leu His Ser Pro Leu His Thr Pro Arg Ala Ser Pro 515 520 525 Thr Gly Thr Pro Gly Thr Thr Pro Pro Pro Ser Pro Gly Gly Gly Val 530 535 540 Gly Gly Ala Ala Trp Arg Ser Arg Leu Asn Ser Ile Arg Asn Ser Phe 545 550 555 560 Leu Gly Ser Pro Arg Phe His Arg Arg Lys Met Gln Val Pro Thr Ala 565 570 575 Glu Glu Met Ser Ser Leu Thr Pro Glu Ser Ser Pro Glu Leu Ala Lys 580 585 590 Arg Ser Trp Phe Gly Asn Phe Ile Ser Leu Asp Lys Glu Glu Gln Ile 595 600 605 Phe Leu Val Leu Lys Asp Lys Pro Leu Ser Ser Ile Lys Ala Asp Ile 610 615 620 Val His Ala Phe Leu Ser Ile Pro Ser Leu Ser His Ser Val Leu Ser 625 630 635 640 Gln Thr Ser Phe Arg Ala Glu Tyr Lys Ala Ser Gly Gly Pro Ser Val 645 650 655 Phe Gln Lys Pro Val Arg Phe Gln Val Asp Ile Ser Ser Ser Glu Gly 660 665 670 Pro Glu Pro Ser Pro Arg Arg Asp Gly Ser Gly Gly Gly Gly Ile Tyr 675 680 685 Ser Val Thr Phe Thr Leu Ile Ser Gly Pro Ser Arg Arg Phe Lys Arg 690 695 700 Val Val Glu Thr Ile Gln Ala Gln Leu Leu Ser Thr His Asp Gln Pro 705 710 715 720 Ser Val Gln Ala Leu Ala Asp Glu Lys Asn Gly Ala Gln Thr Arg Pro 725 730 735 Ala Gly Ala Pro Pro Arg Ser Leu Gln Pro Pro Pro Gly Arg Pro Asp 740 745 750 Pro Glu Leu Ser Ser Ser Pro Arg Arg Gly Pro Pro Lys Asp Lys Lys 755 760 765 Leu Leu Ala Thr Asn Gly Thr Pro Leu Pro 770 775 3 2289 DNA homo sapiens 3 atgggacttg agtttggttt cctagaggct ggtggaaact ggagtcactt cctcccaggg 60 agtgatggga actggaatct cttcctccca ggaattaatg gaaactggag tctcttcctc 120 ccagggaccc atgggaattg gagtctcttt ctcccaagga tcatgggaat tggagttctc 180 tgccaccagg agccagtgga agtgggagac gaggcccttt ggtcctccac atgccccttc 240 cagccctctg ccccctctat atcctttagg tacctggttc tggagcacgt ctcggggggt 300 gagctattcg actacctggt aaagaagggg agactgacgc ccaaggaggc ccgaaagttc 360 ttccgccaga ttgtgtctgc gctggacttc tgccacagct actccatctg ccacagagac 420 ctaaagcccg agaacctgct tttggatgag aaaaacaaca tccgcattgc agacttcggc 480 atggcgtccc tgcaggtggg ggacagcctc ctggagacca gctgcgggtc cccccattat 540 gcgtgtccag aggtgattaa gggggaaaaa tatgatggcc gccgggcaga catgtggagc 600 tgtggagtca tcctcttcgc cctgctcgtg ggggctctgc cctttgatga cgacaacctc 660 cgccagctgc tggagaaggt gaaacggggc gtcttccaca tgccccactt cattcctcca 720 gattgccaga gcctcctgag gggaatgatc gaagtggagc ccgaaaaaag gctcagtctg 780 gagcaaattc agaaacatcc ttggtaccta ggcgggaaac acgagccaga cccgtgcctg 840 gagccagccc ctggccgccg ggtagccatg cggagcctgc catccaacgg agagctggac 900 cccgacgtcc tagagagcat ggcatcactg ggctgcttca gggaccgcga gaggctgcat 960 cgcgagctgc gcagtgagga ggagaaccaa gaaaagatga tatattatct gcttttggat 1020 cggaaggagc ggtatcccag ctgtgaggac caggacctgc ctccccggaa tgatgttgac 1080 cccccccgga agcgtgtgga ttctcccatg ctgagccgtc acgggaagcg gcgaccagag 1140 cggaagtcca tggaagtcct gagcatcacc gatgccgggg gtggtggctc ccctgtaccc 1200 acccgacggg ccttggagat ggcccagcac agccagagat cccgtagcgt cagtggagcc 1260 tccacgggtc tgtcctccag ccctctaagc agcccaagga gtccggtctt ttccttttca 1320 ccggagccgg gggctggaga tgaggctcga ggcgggggct ccccgacttc caaaacgcag 1380 acgctgcctt ctcggggccc caggggtggg ggcgccgggg agcagccccc gccccccagt 1440 gcccgctcca cacccctgcc cggcccccca ggctccccgc gctcctctgg cgggaccccc 1500 ttgcactcgc ctctgcacac gccccgggcc agtcccaccg ggaccccggg gacaacacca 1560 ccccccagcc ccggcggtgg cgtcggggga gccgcctgga ggagtcgtct caactccatc 1620 cgcaacagct tcctgggctc ccctcgcttt caccggcgca agatgcaggt ccctaccgct 1680 gaggagatgt ccagcttgac gccagagtcc tccccggagc tggcaaaacg ctcctggttc 1740 gggaacttca tctccttgga caaagaagaa caaatattcc tcgtgctaaa ggacaaacct 1800 ctcagcagca tcaaagcaga catcgtccat gcctttctgt cgatccccag cctgagtcac 1860 agtgtgctgt cacagaccag cttcagggcc gagtacaagg ccagtggcgg cccctccgtc 1920 ttccaaaagc ccgtccgctt ccaggtggac atcagctcct ctgagggtcc agagccctcc 1980 ccgcgacggg acggcagcgg aggtggtggc atctactccg tcaccttcac tctcatctcg 2040 ggtcccagcc gtcggttcaa gcgagtggtg gagaccatcc aggcacagct cctgagcact 2100 catgaccagc cctccgtgca ggccctggca gacgagaaga acggggccca gacccggcct 2160 gctggtgccc caccccgaag cctgcagccc ccacccggcc gcccagaccc agagctgagc 2220 agctctcccc gccgaggccc ccccaaggac aagaagctcc tggccaccaa cgggacccct 2280 ctgccctga 2289 4 762 PRT homo sapiens 4 Met Gly Leu Glu Phe Gly Phe Leu Glu Ala Gly Gly Asn Trp Ser His 1 5 10 15 Phe Leu Pro Gly Ser Asp Gly Asn Trp Asn Leu Phe Leu Pro Gly Ile 20 25 30 Asn Gly Asn Trp Ser Leu Phe Leu Pro Gly Thr His Gly Asn Trp Ser 35 40 45 Leu Phe Leu Pro Arg Ile Met Gly Ile Gly Val Leu Cys His Gln Glu 50 55 60 Pro Val Glu Val Gly Asp Glu Ala Leu Trp Ser Ser Thr Cys Pro Phe 65 70 75 80 Gln Pro Ser Ala Pro Ser Ile Ser Phe Arg Tyr Leu Val Leu Glu His 85 90 95 Val Ser Gly Gly Glu Leu Phe Asp Tyr Leu Val Lys Lys Gly Arg Leu 100 105 110 Thr Pro Lys Glu Ala Arg Lys Phe Phe Arg Gln Ile Val Ser Ala Leu 115 120 125 Asp Phe Cys His Ser Tyr Ser Ile Cys His Arg Asp Leu Lys Pro Glu 130 135 140 Asn Leu Leu Leu Asp Glu Lys Asn Asn Ile Arg Ile Ala Asp Phe Gly 145 150 155 160 Met Ala Ser Leu Gln Val Gly Asp Ser Leu Leu Glu Thr Ser Cys Gly 165 170 175 Ser Pro His Tyr Ala Cys Pro Glu Val Ile Lys Gly Glu Lys Tyr Asp 180 185 190 Gly Arg Arg Ala Asp Met Trp Ser Cys Gly Val Ile Leu Phe Ala Leu 195 200 205 Leu Val Gly Ala Leu Pro Phe Asp Asp Asp Asn Leu Arg Gln Leu Leu 210 215 220 Glu Lys Val Lys Arg Gly Val Phe His Met Pro His Phe Ile Pro Pro 225 230 235 240 Asp Cys Gln Ser Leu Leu Arg Gly Met Ile Glu Val Glu Pro Glu Lys 245 250 255 Arg Leu Ser Leu Glu Gln Ile Gln Lys His Pro Trp Tyr Leu Gly Gly 260 265 270 Lys His Glu Pro Asp Pro Cys Leu Glu Pro Ala Pro Gly Arg Arg Val 275 280 285 Ala Met Arg Ser Leu Pro Ser Asn Gly Glu Leu Asp Pro Asp Val Leu 290 295 300 Glu Ser Met Ala Ser Leu Gly Cys Phe Arg Asp Arg Glu Arg Leu His 305 310 315 320 Arg Glu Leu Arg Ser Glu Glu Glu Asn Gln Glu Lys Met Ile Tyr Tyr 325 330 335 Leu Leu Leu Asp Arg Lys Glu Arg Tyr Pro Ser Cys Glu Asp Gln Asp 340 345 350 Leu Pro Pro Arg Asn Asp Val Asp Pro Pro Arg Lys Arg Val Asp Ser 355 360 365 Pro Met Leu Ser Arg His Gly Lys Arg Arg Pro Glu Arg Lys Ser Met 370 375 380 Glu Val Leu Ser Ile Thr Asp Ala Gly Gly Gly Gly Ser Pro Val Pro 385 390 395 400 Thr Arg Arg Ala Leu Glu Met Ala Gln His Ser Gln Arg Ser Arg Ser 405 410 415 Val Ser Gly Ala Ser Thr Gly Leu Ser Ser Ser Pro Leu Ser Ser Pro 420 425 430 Arg Ser Pro Val Phe Ser Phe Ser Pro Glu Pro Gly Ala Gly Asp Glu 435 440 445 Ala Arg Gly Gly Gly Ser Pro Thr Ser Lys Thr Gln Thr Leu Pro Ser 450 455 460 Arg Gly Pro Arg Gly Gly Gly Ala Gly Glu Gln Pro Pro Pro Pro Ser 465 470 475 480 Ala Arg Ser Thr Pro Leu Pro Gly Pro Pro Gly Ser Pro Arg Ser Ser 485 490 495 Gly Gly Thr Pro Leu His Ser Pro Leu His Thr Pro Arg Ala Ser Pro 500 505 510 Thr Gly Thr Pro Gly Thr Thr Pro Pro Pro Ser Pro Gly Gly Gly Val 515 520 525 Gly Gly Ala Ala Trp Arg Ser Arg Leu Asn Ser Ile Arg Asn Ser Phe 530 535 540 Leu Gly Ser Pro Arg Phe His Arg Arg Lys Met Gln Val Pro Thr Ala 545 550 555 560 Glu Glu Met Ser Ser Leu Thr Pro Glu Ser Ser Pro Glu Leu Ala Lys 565 570 575 Arg Ser Trp Phe Gly Asn Phe Ile Ser Leu Asp Lys Glu Glu Gln Ile 580 585 590 Phe Leu Val Leu Lys Asp Lys Pro Leu Ser Ser Ile Lys Ala Asp Ile 595 600 605 Val His Ala Phe Leu Ser Ile Pro Ser Leu Ser His Ser Val Leu Ser 610 615 620 Gln Thr Ser Phe Arg Ala Glu Tyr Lys Ala Ser Gly Gly Pro Ser Val 625 630 635 640 Phe Gln Lys Pro Val Arg Phe Gln Val Asp Ile Ser Ser Ser Glu Gly 645 650 655 Pro Glu Pro Ser Pro Arg Arg Asp Gly Ser Gly Gly Gly Gly Ile Tyr 660 665 670 Ser Val Thr Phe Thr Leu Ile Ser Gly Pro Ser Arg Arg Phe Lys Arg 675 680 685 Val Val Glu Thr Ile Gln Ala Gln Leu Leu Ser Thr His Asp Gln Pro 690 695 700 Ser Val Gln Ala Leu Ala Asp Glu Lys Asn Gly Ala Gln Thr Arg Pro 705 710 715 720 Ala Gly Ala Pro Pro Arg Ser Leu Gln Pro Pro Pro Gly Arg Pro Asp 725 730 735 Pro Glu Leu Ser Ser Ser Pro Arg Arg Gly Pro Pro Lys Asp Lys Lys 740 745 750 Leu Leu Ala Thr Asn Gly Thr Pro Leu Pro 755 760 5 2112 DNA homo sapiens 5 atgaaggtgg agcgggagat cgccatcctg aagctcatcg aacacccaca tgtcctcaag 60 ctccacgacg tctacgagaa caagaaatat ttgtacctgg ttctggagca cgtctcgggg 120 ggtgagctat tcgactacct ggtaaagaag gggagactga cgcccaagga ggcccgaaag 180 ttcttccgcc agattgtgtc tgcgctggac ttctgccaca gctactccat ctgccacaga 240 gacctaaagc ccgagaacct gcttttggat gagaaaaaca acatccgcat tgcagacttc 300 ggcatggcgt ccctgcaggt gggggacagc ctcctggaga ccagctgcgg gtccccccat 360 tatgcgtgtc cagaggtgat taagggggaa aaatatgatg gccgccgggc agacatgtgg 420 agctgtggag tcatcctctt cgccctgctc gtgggggctc tgccctttga tgacgacaac 480 ctccgccagc tgctggagaa ggtgaaacgg ggcgtcttcc acatgcccca cttcattcct 540 ccagattgcc agagcctcct gaggggaatg atcgaagtgg agcccgaaaa aaggctcagt 600 ctggagcaaa ttcagaaaca tccttggtac ctaggcggga aacacgagcc agacccgtgc 660 ctggagccag cccctggccg ccgggtagcc atgcggagcc tgccatccaa cggagagctg 720 gaccccgacg tcctagagag catggcatca ctgggctgct tcagggaccg cgagaggctg 780 catcgcgagc tgcgcagtga ggaggagaac caagaaaaga tgatatatta tctgcttttg 840 gatcggaagg agcggtatcc cagctgtgag gaccaggacc tgcctccccg gaatgatgtt 900 gacccccccc ggaagcgtgt ggattctccc atgctgagcc gtcacgggaa gcggcgacca 960 gagcggaagt ccatggaagt cctgagcatc accgatgccg ggggtggtgg ctcccctgta 1020 cccacccgac gggccttgga gatggcccag cacagccaga gatcccgtag cgtcagtgga 1080 gcctccacgg gtctgtcctc cagccctcta agcagcccaa ggagtccggt cttttccttt 1140 tcaccggagc cgggggctgg agatgaggct cgaggcgggg gctccccgac ttccaaaacg 1200 cagacgctgc cttctcgggg ccccaggggt gggggcgccg gggagcagcc cccgcccccc 1260 agtgcccgct ccacacccct gcccggcccc ccaggctccc cgcgctcctc tggcgggacc 1320 cccttgcact cgcctctgca cacgccccgg gccagtccca ccgggacccc ggggacaaca 1380 ccacccccca gccccggcgg tggcgtcggg ggagccgcct ggaggagtcg tctcaactcc 1440 atccgcaaca gcttcctggg ctcccctcgc tttcaccggc gcaagatgca ggtccctacc 1500 gctgaggaga tgtccagctt gacgccagag tcctccccgg agctggcaaa acgctcctgg 1560 ttcgggaact tcatctcctt ggacaaagaa gaacaaatat tcctcgtgct aaaggacaaa 1620 cctctcagca gcatcaaagc agacatcgtc catgcctttc tgtcgatccc cagcctgagt 1680 cacagtgtgc tgtcacagac cagcttcagg gccgagtaca aggccagtgg cggcccctcc 1740 gtcttccaaa agcccgtccg cttccaggtg gacatcagct cctctgaggg tccagagccc 1800 tccccgcgac gggacggcag cggaggtggt ggcatctact ccgtcacctt cactctcatc 1860 tcgggtccca gccgtcggtt caagcgagtg gtggagacca tccaggcaca gctcctgagc 1920 actcatgacc agccctccgt gcaggccctg gcagacgaga agaacggggc ccagacccgg 1980 cctgctggtg ccccaccccg aagcctgcag cccccacccg gccgcccaga cccagagctg 2040 agcagctctc cccgccgagg cccccccaag gacaagaagc tcctggccac caacgggacc 2100 cctctgccct ga 2112 6 703 PRT homo sapiens 6 Met Lys Val Glu Arg Glu Ile Ala Ile Leu Lys Leu Ile Glu His Pro 1 5 10 15 His Val Leu Lys Leu His Asp Val Tyr Glu Asn Lys Lys Tyr Leu Tyr 20 25 30 Leu Val Leu Glu His Val Ser Gly Gly Glu Leu Phe Asp Tyr Leu Val 35 40 45 Lys Lys Gly Arg Leu Thr Pro Lys Glu Ala Arg Lys Phe Phe Arg Gln 50 55 60 Ile Val Ser Ala Leu Asp Phe Cys His Ser Tyr Ser Ile Cys His Arg 65 70 75 80 Asp Leu Lys Pro Glu Asn Leu Leu Leu Asp Glu Lys Asn Asn Ile Arg 85 90 95 Ile Ala Asp Phe Gly Met Ala Ser Leu Gln Val Gly Asp Ser Leu Leu 100 105 110 Glu Thr Ser Cys Gly Ser Pro His Tyr Ala Cys Pro Glu Val Ile Lys 115 120 125 Gly Glu Lys Tyr Asp Gly Arg Arg Ala Asp Met Trp Ser Cys Gly Val 130 135 140 Ile Leu Phe Ala Leu Leu Val Gly Ala Leu Pro Phe Asp Asp Asp Asn 145 150 155 160 Leu Arg Gln Leu Leu Glu Lys Val Lys Arg Gly Val Phe His Met Pro 165 170 175 His Phe Ile Pro Pro Asp Cys Gln Ser Leu Leu Arg Gly Met Ile Glu 180 185 190 Val Glu Pro Glu Lys Arg Leu Ser Leu Glu Gln Ile Gln Lys His Pro 195 200 205 Trp Tyr Leu Gly Gly Lys His Glu Pro Asp Pro Cys Leu Glu Pro Ala 210 215 220 Pro Gly Arg Arg Val Ala Met Arg Ser Leu Pro Ser Asn Gly Glu Leu 225 230 235 240 Asp Pro Asp Val Leu Glu Ser Met Ala Ser Leu Gly Cys Phe Arg Asp 245 250 255 Arg Glu Arg Leu His Arg Glu Leu Arg Ser Glu Glu Glu Asn Gln Glu 260 265 270 Lys Met Ile Tyr Tyr Leu Leu Leu Asp Arg Lys Glu Arg Tyr Pro Ser 275 280 285 Cys Glu Asp Gln Asp Leu Pro Pro Arg Asn Asp Val Asp Pro Pro Arg 290 295 300 Lys Arg Val Asp Ser Pro Met Leu Ser Arg His Gly Lys Arg Arg Pro 305 310 315 320 Glu Arg Lys Ser Met Glu Val Leu Ser Ile Thr Asp Ala Gly Gly Gly 325 330 335 Gly Ser Pro Val Pro Thr Arg Arg Ala Leu Glu Met Ala Gln His Ser 340 345 350 Gln Arg Ser Arg Ser Val Ser Gly Ala Ser Thr Gly Leu Ser Ser Ser 355 360 365 Pro Leu Ser Ser Pro Arg Ser Pro Val Phe Ser Phe Ser Pro Glu Pro 370 375 380 Gly Ala Gly Asp Glu Ala Arg Gly Gly Gly Ser Pro Thr Ser Lys Thr 385 390 395 400 Gln Thr Leu Pro Ser Arg Gly Pro Arg Gly Gly Gly Ala Gly Glu Gln 405 410 415 Pro Pro Pro Pro Ser Ala Arg Ser Thr Pro Leu Pro Gly Pro Pro Gly 420 425 430 Ser Pro Arg Ser Ser Gly Gly Thr Pro Leu His Ser Pro Leu His Thr 435 440 445 Pro Arg Ala Ser Pro Thr Gly Thr Pro Gly Thr Thr Pro Pro Pro Ser 450 455 460 Pro Gly Gly Gly Val Gly Gly Ala Ala Trp Arg Ser Arg Leu Asn Ser 465 470 475 480 Ile Arg Asn Ser Phe Leu Gly Ser Pro Arg Phe His Arg Arg Lys Met 485 490 495 Gln Val Pro Thr Ala Glu Glu Met Ser Ser Leu Thr Pro Glu Ser Ser 500 505 510 Pro Glu Leu Ala Lys Arg Ser Trp Phe Gly Asn Phe Ile Ser Leu Asp 515 520 525 Lys Glu Glu Gln Ile Phe Leu Val Leu Lys Asp Lys Pro Leu Ser Ser 530 535 540 Ile Lys Ala Asp Ile Val His Ala Phe Leu Ser Ile Pro Ser Leu Ser 545 550 555 560 His Ser Val Leu Ser Gln Thr Ser Phe Arg Ala Glu Tyr Lys Ala Ser 565 570 575 Gly Gly Pro Ser Val Phe Gln Lys Pro Val Arg Phe Gln Val Asp Ile 580 585 590 Ser Ser Ser Glu Gly Pro Glu Pro Ser Pro Arg Arg Asp Gly Ser Gly 595 600 605 Gly Gly Gly Ile Tyr Ser Val Thr Phe Thr Leu Ile Ser Gly Pro Ser 610 615 620 Arg Arg Phe Lys Arg Val Val Glu Thr Ile Gln Ala Gln Leu Leu Ser 625 630 635 640 Thr His Asp Gln Pro Ser Val Gln Ala Leu Ala Asp Glu Lys Asn Gly 645 650 655 Ala Gln Thr Arg Pro Ala Gly Ala Pro Pro Arg Ser Leu Gln Pro Pro 660 665 670 Pro Gly Arg Pro Asp Pro Glu Leu Ser Ser Ser Pro Arg Arg Gly Pro 675 680 685 Pro Lys Asp Lys Lys Leu Leu Ala Thr Asn Gly Thr Pro Leu Pro 690 695 700

Claims (5)

What is claimed is:
1. An isolated nucleic acid molecule comprising a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6.
2. The isolated nucleic acid molecule of claim 1, comprising a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO:2.
3. The isolated nucleic acid molecule of claim 1, comprising a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO:4.
4. The isolated nucleic acid molecule of claim 1, comprising a nucleotide sequence encoding the amino acid sequence shown in SEQ ID NO:6.
5. A substantially isolated protein having the kinase activity of the protein shown in SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:6, which is encoded by a nucleotide sequence that hybridizes to SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:5 under highly stringent conditions.
US10/803,277 1999-09-28 2004-03-18 Human kinases and polynucleotides encoding the same Expired - Fee Related US6919192B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/803,277 US6919192B2 (en) 2001-04-06 2004-03-18 Human kinases and polynucleotides encoding the same
US11/165,424 US20080050809A1 (en) 1999-09-28 2005-06-23 Novel human kinases and polynucleotides encoding the same

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US28203601P 2001-04-06 2001-04-06
US10/116,326 US6777545B2 (en) 2001-04-06 2002-04-04 Human kinases and polynucleotides encoding the same
US10/803,277 US6919192B2 (en) 2001-04-06 2004-03-18 Human kinases and polynucleotides encoding the same

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/116,326 Division US6777545B2 (en) 1999-09-28 2002-04-04 Human kinases and polynucleotides encoding the same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/103,547 Continuation-In-Part US6759527B2 (en) 1999-09-28 2002-03-20 Human kinase and polynucleotides encoding the same

Publications (2)

Publication Number Publication Date
US20040180416A1 true US20040180416A1 (en) 2004-09-16
US6919192B2 US6919192B2 (en) 2005-07-19

Family

ID=23079830

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/116,326 Expired - Fee Related US6777545B2 (en) 1999-09-28 2002-04-04 Human kinases and polynucleotides encoding the same
US10/803,277 Expired - Fee Related US6919192B2 (en) 1999-09-28 2004-03-18 Human kinases and polynucleotides encoding the same

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/116,326 Expired - Fee Related US6777545B2 (en) 1999-09-28 2002-04-04 Human kinases and polynucleotides encoding the same

Country Status (5)

Country Link
US (2) US6777545B2 (en)
EP (1) EP1383879A4 (en)
JP (1) JP2004524043A (en)
CA (1) CA2443516A1 (en)
WO (1) WO2002081670A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080050809A1 (en) * 1999-09-28 2008-02-28 Alejandro Abuin Novel human kinases and polynucleotides encoding the same
EP1396545A4 (en) * 2001-06-04 2004-12-08 Taiho Pharmaceutical Co Ltd Cell cycle regulatory factor
AU2002352774A1 (en) * 2001-11-15 2003-05-26 Millennium Pharmaceuticals, Inc. 55053, a novel human eukaryotic kinase and uses therefor

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4215051A (en) * 1979-08-29 1980-07-29 Standard Oil Company (Indiana) Formation, purification and recovery of phthalic anhydride
US4376110A (en) * 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4594595A (en) * 1984-04-18 1986-06-10 Sanders Associates, Inc. Circular log-periodic direction-finder array
US4631211A (en) * 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
US4689405A (en) * 1983-01-20 1987-08-25 Gesellschaft Fur Biotechnologische Forschung Mbh (Gbf) Process for the simultaneous synthesis of several oligonucleotides on a solid phase
US4713326A (en) * 1983-07-05 1987-12-15 Molecular Diagnostics, Inc. Coupling of nucleic acids to solid support by photochemical methods
US4873191A (en) * 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US5272057A (en) * 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5424186A (en) * 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5445934A (en) * 1989-06-07 1995-08-29 Affymax Technologies N.V. Array of oligonucleotides on a solid substrate
US5459127A (en) * 1990-04-19 1995-10-17 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5556752A (en) * 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5700637A (en) * 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5723323A (en) * 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US5744305A (en) * 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5756289A (en) * 1991-07-03 1998-05-26 Salk Institute For Biological Studies Protein kinases
US5817479A (en) * 1996-08-07 1998-10-06 Incyte Pharmaceuticals, Inc. Human kinase homologs
US5830721A (en) * 1994-02-17 1998-11-03 Affymax Technologies N.V. DNA mutagenesis by random fragmentation and reassembly
US5837458A (en) * 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5869336A (en) * 1994-07-15 1999-02-09 Cephalon, Inc. Recombinant enzymatically active calpain expressed in a baculovirus system
US5877397A (en) * 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US6075181A (en) * 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6110490A (en) * 1994-08-05 2000-08-29 The United States Of America As Represented By The Department Of Health And Human Services Liposomal delivery system for biologically active agents
US6114598A (en) * 1990-01-12 2000-09-05 Abgenix, Inc. Generation of xenogeneic antibodies
US6117679A (en) * 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US6150584A (en) * 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6787345B1 (en) * 2000-11-15 2004-09-07 Millennium Pharmaceuticals, Inc. 55053, a novel human eukaryotic kinase and uses therefor

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003514583A (en) * 1999-11-24 2003-04-22 スージェン・インコーポレーテッド Novel human protein kinases and protein kinase-like enzymes

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4215051A (en) * 1979-08-29 1980-07-29 Standard Oil Company (Indiana) Formation, purification and recovery of phthalic anhydride
US4376110A (en) * 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4873191A (en) * 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4689405A (en) * 1983-01-20 1987-08-25 Gesellschaft Fur Biotechnologische Forschung Mbh (Gbf) Process for the simultaneous synthesis of several oligonucleotides on a solid phase
US4713326A (en) * 1983-07-05 1987-12-15 Molecular Diagnostics, Inc. Coupling of nucleic acids to solid support by photochemical methods
US4594595A (en) * 1984-04-18 1986-06-10 Sanders Associates, Inc. Circular log-periodic direction-finder array
US4631211A (en) * 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
US5723323A (en) * 1985-03-30 1998-03-03 Kauffman; Stuart Alan Method of identifying a stochastically-generated peptide, polypeptide, or protein having ligand binding property and compositions thereof
US4946778A (en) * 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5700637A (en) * 1988-05-03 1997-12-23 Isis Innovation Limited Apparatus and method for analyzing polynucleotide sequences and method of generating oligonucleotide arrays
US5272057A (en) * 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5445934A (en) * 1989-06-07 1995-08-29 Affymax Technologies N.V. Array of oligonucleotides on a solid substrate
US5744305A (en) * 1989-06-07 1998-04-28 Affymetrix, Inc. Arrays of materials attached to a substrate
US5424186A (en) * 1989-06-07 1995-06-13 Affymax Technologies N.V. Very large scale immobilized polymer synthesis
US5252743A (en) * 1989-11-13 1993-10-12 Affymax Technologies N.V. Spatially-addressable immobilization of anti-ligands on surfaces
US6150584A (en) * 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6114598A (en) * 1990-01-12 2000-09-05 Abgenix, Inc. Generation of xenogeneic antibodies
US6075181A (en) * 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5459127A (en) * 1990-04-19 1995-10-17 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5877397A (en) * 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5756289A (en) * 1991-07-03 1998-05-26 Salk Institute For Biological Studies Protein kinases
US5837458A (en) * 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5830721A (en) * 1994-02-17 1998-11-03 Affymax Technologies N.V. DNA mutagenesis by random fragmentation and reassembly
US6117679A (en) * 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US5869336A (en) * 1994-07-15 1999-02-09 Cephalon, Inc. Recombinant enzymatically active calpain expressed in a baculovirus system
US6110490A (en) * 1994-08-05 2000-08-29 The United States Of America As Represented By The Department Of Health And Human Services Liposomal delivery system for biologically active agents
US5556752A (en) * 1994-10-24 1996-09-17 Affymetrix, Inc. Surface-bound, unimolecular, double-stranded DNA
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5817479A (en) * 1996-08-07 1998-10-06 Incyte Pharmaceuticals, Inc. Human kinase homologs
US6787345B1 (en) * 2000-11-15 2004-09-07 Millennium Pharmaceuticals, Inc. 55053, a novel human eukaryotic kinase and uses therefor

Also Published As

Publication number Publication date
US20030166889A1 (en) 2003-09-04
EP1383879A1 (en) 2004-01-28
US6919192B2 (en) 2005-07-19
JP2004524043A (en) 2004-08-12
US6777545B2 (en) 2004-08-17
CA2443516A1 (en) 2002-10-17
EP1383879A4 (en) 2005-05-11
WO2002081670A1 (en) 2002-10-17

Similar Documents

Publication Publication Date Title
US6777545B2 (en) Human kinases and polynucleotides encoding the same
US6476210B2 (en) Human kinases and polynucleotides encoding the same
US20050266452A1 (en) Novel human kinases and polynucleotides encoding the same
US6734009B2 (en) Human kinases and polynucleotides encoding the same
US6617147B2 (en) Human kinase proteins and polynucleotides encoding the same
US6734010B2 (en) Human kinases and polynucleotides encoding the same
US6849443B2 (en) Human kinases and polynucleotides encoding the same
CA2431007A1 (en) Novel human kinase and polynucleotides encoding the same
US6586230B1 (en) Human kinase and polynucleotides encoding the same
US6797510B1 (en) Human kinases and polynucleotides encoding the same
US20020147320A1 (en) Novel human kinase proteins and polynucleotides encoding the same
US6815188B2 (en) Human kinases and polynucleotides encoding the same
AU2002303254A1 (en) Novel human kinases and polynucleotides encoding the same
AU2002243634A1 (en) Novel human kinases and polynucleotides encoding the same

Legal Events

Date Code Title Description
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20090719